88 results on '"Brendan D. Price"'
Search Results
2. Supplementary Figures S1-S4 from Autophagy Induction with RAD001 Enhances Chemosensitivity and Radiosensitivity through Met Inhibition in Papillary Thyroid Cancer
- Author
-
Daniel T. Ruan, Francis D. Moore, Brendan D. Price, Xiaofeng Jiang, Frank He, Jochen Lorch, Jinyan Du, David B. Donner, Edward E. Whang, and Chi-Iou Lin
- Abstract
Supplementary Figures S1-S4.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Galectin-3 Targeted Therapy with a Small Molecule Inhibitor Activates Apoptosis and Enhances Both Chemosensitivity and Radiosensitivity in Papillary Thyroid Cancer
- Author
-
Daniel T. Ruan, Francis D. Moore, Vania Nose, Ulf J. Nilsson, Hakon Leffler, Tamara Delaine, Adelaide M. Carothers, Brendan D. Price, Xiaofeng Jiang, David B. Donner, Edward E. Whang, and Chi-Iou Lin
- Abstract
Although most patients with papillary thyroid cancer (PTC) have favorable outcomes, some have advanced PTC that is refractory to external beam radiation and systemic chemotherapy. Galectin-3 (Gal-3) is a β-galactoside–binding protein with antiapoptotic activity that is consistently overexpressed in PTC. The purpose of this study is to determine if Gal-3 inhibition promotes apoptosis, chemosensitivity, and radiosensitivity in PTC. PTC cell lines (8505-C and TPC-1) and human ex vivo PTC were treated with a highly specific small molecule inhibitor of Gal-3 (Td131_1). Apoptotic activity was determined by flow cytometric analysis as well as caspase-3 and PARP cleavage. The minimum inhibitory concentrations of Td131_1 and doxorubicin were determined, and their combined effects were measured to test for synergistic activity. The effects of Td131_1 on radiosensitivity were determined by a clonogenic assay. Td131_1 promoted apoptosis, improved radiosensitivity, and synergistically enhanced chemosensitivity to doxorubicin in PTC cell lines. In PTC ex vivo, Td131_1 treatment alone induced the cleavage of caspase-3 and PARP. Td131_1 and doxorubicin together activated apoptosis in PTC ex vivo to a greater degree than their combined individual effects. Td131_1 activated apoptosis and had synergistic activity with doxorubicin in PTC. We conclude that Gal-3 targeted therapy is a promising therapeutic strategy for advanced PTC that is refractory to surgery and radioactive iodine therapy. (Mol Cancer Res 2009;7(10):1655–62)
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Data from High-Throughput Screening Identifies Two Classes of Antibiotics as Radioprotectors: Tetracyclines and Fluoroquinolones
- Author
-
William H. McBride, Richard A. Gatti, Brendan D. Price, James W. Sayre, Keisuke S. Iwamoto, Robert Damoiseaux, Kelly Pettijohn, Ewa Micewicz, Yingli Sun, J. Tyson McDonald, Andrew J. Norris, Julianne M. Pollard, and Kwanghee Kim
- Abstract
Supplementary Data from High-Throughput Screening Identifies Two Classes of Antibiotics as Radioprotectors: Tetracyclines and Fluoroquinolones
- Published
- 2023
- Full Text
- View/download PDF
5. Polymerase δ promotes chromosomal rearrangements and imprecise double-strand break repair
- Author
-
Michael T. Hemann, Alexandria Van Scoyk, Jacob V. Layer, Alexander J. Brown, Tovah A. Day, Yunpeng Liu, Brendan D. Price, Steven A. Roberts, Kristen E. Stevenson, Nealia C.M. House, David M. Weinstock, and Lydie Debaize
- Subjects
chemistry.chemical_classification ,Exonuclease ,DNA ligase ,DNA End-Joining Repair ,Multidisciplinary ,POLD1 ,biology ,Chemistry ,DNA polymerase ,LIG3 ,Biological Sciences ,Translocation, Genetic ,Double Strand Break Repair ,Cell biology ,Non-homologous end joining ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,Gene Knockdown Techniques ,biology.protein ,Humans ,DNA Breaks, Double-Stranded ,RNA, Small Interfering ,Polymerase ,DNA Polymerase III ,HeLa Cells - Abstract
Recent studies have implicated DNA polymerases θ (Pol θ) and β (Pol β) as mediators of alternative nonhomologous end-joining (Alt-NHEJ) events, including chromosomal translocations. Here we identify subunits of the replicative DNA polymerase δ (Pol δ) as promoters of Alt-NHEJ that results in more extensive intrachromosomal mutations at a single double-strand break (DSB) and more frequent translocations between two DSBs. Depletion of the Pol δ accessory subunit POLD2 destabilizes the complex, resulting in degradation of both POLD1 and POLD3 in human cells. POLD2 depletion markedly reduces the frequency of translocations with sequence modifications but does not affect the frequency of translocations with exact joins. Using separation-of-function mutants, we show that both the DNA synthesis and exonuclease activities of the POLD1 subunit contribute to translocations. As described in yeast and unlike Pol θ, Pol δ also promotes homology-directed repair. Codepletion of POLD2 with 53BP1 nearly eliminates translocations. POLD1 and POLD2 each colocalize with phosphorylated H2AX at ionizing radiation-induced DSBs but not with 53BP1. Codepletion of POLD2 with either ligase 3 (LIG3) or ligase 4 (LIG4) does not further reduce translocation frequency compared to POLD2 depletion alone. Together, these data support a model in which Pol δ promotes Alt-NHEJ in human cells at DSBs, including translocations.
- Published
- 2020
- Full Text
- View/download PDF
6. The ZEB2-dependent EMT transcriptional programme drives therapy resistance by activating nucleotide excision repair genes ERCC1 and ERCC4 in colorectal cancer
- Author
-
Eugene Tulchinsky, Tamer Yagci, Seval Kilic, Maria Antonette Lopez, Rahul Sreekumar, Brendan D. Price, Metin Çetin, Muhammad Emaduddin, Ashish Patel, Sule Erdemir, Karwan A. Moutasim, Hajir Al-Saihati, Marta Salgado Navio, A. Emre Sayan, John N. Primrose, Geert Berx, Nathan Curtis, Gareth J. Thomas, Massimiliano Mellone, and Alex H. Mirnezami
- Subjects
0301 basic medicine ,Cancer Research ,DNA Repair ,Organoplatinum Compounds ,Transcription, Genetic ,Colorectal cancer ,TO-MESENCHYMAL TRANSITION ,Leucovorin ,Mice ,0302 clinical medicine ,FOLFOX ,Antineoplastic Combined Chemotherapy Protocols ,Medicine and Health Sciences ,ZEB1 ,RC254-282 ,Research Articles ,ZEB2 ,CHEMORESISTANCE ,MOLECULAR-MECHANISMS ,Liver Neoplasms ,EMT ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,General Medicine ,CHEMOTHERAPY ,DNA-Binding Proteins ,Oncology ,030220 oncology & carcinogenesis ,DNA-REPAIR ,Molecular Medicine ,Biomarker (medicine) ,Fluorouracil ,Colorectal Neoplasms ,medicine.drug ,Research Article ,EXPRESSION ,Epithelial-Mesenchymal Transition ,PROTEINS ,DNA repair ,03 medical and health sciences ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Epithelial–mesenchymal transition ,neoplasms ,Zinc Finger E-box Binding Homeobox 2 ,business.industry ,oxaliplatin ,Biology and Life Sciences ,BLADDER ,medicine.disease ,Endonucleases ,Xenograft Model Antitumor Assays ,digestive system diseases ,Oxaliplatin ,030104 developmental biology ,Drug Resistance, Neoplasm ,CELLS ,Cancer research ,ERCC1 ,business ,ERCC4 ,Nucleotide excision repair - Abstract
Resistance to adjuvant chemotherapy is a major clinical problem in the treatment of colorectal cancer (CRC). The aim of this study was to elucidate the role of an epithelial to mesenchymal transition (EMT)‐inducing protein, ZEB2, in chemoresistance of CRC, and to uncover the underlying mechanism. We performed IHC for ZEB2 and association analyses with clinical outcomes on primary CRC and matched CRC liver metastases in compliance with observational biomarker study guidelines. ZEB2 expression in primary tumours was an independent prognostic marker of reduced overall survival and disease‐free survival in patients who received adjuvant FOLFOX chemotherapy. ZEB2 expression was retained in 96% of liver metastases. The ZEB2‐dependent EMT transcriptional programme activated nucleotide excision repair (NER) pathway largely via upregulation of the ERCC1 gene and other components in NER pathway, leading to enhanced viability of CRC cells upon oxaliplatin treatment. ERCC1‐overexpressing CRC cells did not respond to oxaliplatin in vivo, as assessed using a murine orthotopic model in a randomised and blinded preclinical study. Our findings show that ZEB2 is a biomarker of tumour response to chemotherapy and risk of recurrence in CRC patients. We propose that the ZEB2‐ERCC1 axis is a key determinant of chemoresistance in CRC., Here, we show that the expression of ZEB2 marks reduced overall and disease‐free survival in primary and secondary colorectal cancers (CRCs). ZEB2 overexpression promoted chemoresistance to oxaliplatin in vitro and in vivo by transcriptionally activating nucleotide excision repair genes ERCC1 and ERCC4. Overall ZEB2 is a promising biomarker predicting both therapy response and metastatic ability of CRCs.
- Published
- 2021
7. Site-specific targeting of a light activated dCas9-KIllerRed fusion protein generates transient, localized regions of oxidative DNA damage
- Author
-
Jacob V. Layer, Nealia C.M. House, and Brendan D. Price
- Subjects
Ku70 ,chemistry.chemical_compound ,DNA damage ,Cas9 ,DNA repair ,Chemistry ,CRISPR ,Fusion protein ,DNA ,Cell biology ,Chromatin - Abstract
DNA repair requires reorganization of the local chromatin structure to facilitate access to and repair of the DNA. Studying DNA double-strand break (DSB) repair in specific chromatin domains has been aided by the use of sequence-specific endonucleases to generate targeted breaks. Here, we describe a new approach that combines KillerRed, a photosensitizer that generates reactive oxygen species (ROS) when exposed to light, and the genome-targeting properties of the CRISPR/Cas9 system. Fusing KillerRed to catalytically inactive Cas9 (dCas9) generates dCas9-KR, which can then be targeted to any desired genomic region with an appropriate guide RNA. Activation of dCas9-KR with green light generates a local increase in reactive oxygen species, resulting in “clustered” oxidative damage, including both DNA breaks and base damage. Activation of dCas9-KR rapidly (within minutes) increases both γH2AX and recruitment of the KU70/80 complex. Importantly, this damage is repaired within 10 minutes of termination of light exposure, indicating that the DNA damage generated by dCas9-KR is both rapid and transient. Further, repair is carried out exclusively through NHEJ, with no detectable contribution from HR-based mechanisms. Surprisingly, sequencing of repaired DNA damage regions did not reveal any increase in either mutations or INDELs in the targeted region, implying that NHEJ has high fidelity under the conditions of low level, limited damage. The dCas9-KR approach for creating targeted damage has significant advantages over the use of endonucleases, since the duration and intensity of DNA damage can be controlled in “real time” by controlling light exposure. In addition, unlike endonucleases that carry out multiple cut-repair cycles, dCas9-KR produces a single burst of damage, more closely resembling the type of damage experienced during acute exposure to reactive oxygen species or environmental toxins. dCas9-KR is a promising system to induce DNA damage and measure site-specific repair kinetics at clustered DNA lesions.
- Published
- 2020
- Full Text
- View/download PDF
8. Multiple Roles for Mono- and Poly(ADP-Ribose) in Regulating Stress Responses
- Author
-
Hongyun Qi, Brendan D. Price, and Tovah A. Day
- Subjects
Poly Adenosine Diphosphate Ribose ,DNA damage ,Poly ADP ribose polymerase ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,ADP-Ribosylation ,0302 clinical medicine ,Protein Domains ,Stress, Physiological ,Cellular stress response ,Ribose ,Genetics ,Polymerase ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,Proteins ,Metabolism ,Cell biology ,Enzyme ,chemistry ,biology.protein ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery ,Function (biology) ,DNA Damage ,Signal Transduction - Abstract
Although stress-induced synthesis of mono(ADP-ribose) (mADPr) and poly(ADP-ribose) (pADPr) conjugates by pADPr polymerase (PARP) enzymes has been studied extensively, the removal and degradation of pADPr, as well as the fate of ADPr metabolites, have received less attention. The observations that stress-induced pADPr undergoes rapid turnover, and that deficiencies in ADPr degradation phenocopy loss of pADPr synthesis, suggest that ADPr degradation is fundamentally important to the cellular stress response. Recent work has identified several distinct families of pADPr hydrolases that can degrade pADPr to release pADPr or mADPr into the cytoplasm. Further, many stress-response proteins contain ADPr-binding domains that can interact with these metabolites. We discuss how pADPr metabolites generated during pADPr degradation can function as signaling intermediates in processes such as inflammation, apoptosis, and DNA damage responses. These studies highlight that the full cycle of ADPr metabolism, including both synthesis and degradation, is necessary for responses to genotoxic stress.
- Published
- 2019
- Full Text
- View/download PDF
9. PARP3 is a promoter of chromosomal rearrangements and limits G4 DNA
- Author
-
William C. Hahn, David E. Root, Anna C. Schinzel, J. Patrick Cleary, David M. Weinstock, Brendan D. Price, Srijoy Guha, John G. Doench, Jacob V. Layer, Tovah A. Day, Trevor Tivey, Kristen E. Stevenson, Sunhee Kim, and Francesca Izzo
- Subjects
0301 basic medicine ,Multidisciplinary ,biology ,DNA damage ,DNA repair ,Poly ADP ribose polymerase ,Science ,General Physics and Astronomy ,General Chemistry ,Molecular biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,DNA End-Joining Repair ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,Rad50 ,biology.protein ,Homologous recombination ,Polymerase ,DNA - Abstract
Chromosomal rearrangements are essential events in the pathogenesis of both malignant and nonmalignant disorders, yet the factors affecting their formation are incompletely understood. Here we develop a zinc-finger nuclease translocation reporter and screen for factors that modulate rearrangements in human cells. We identify UBC9 and RAD50 as suppressors and 53BP1, DDB1 and poly(ADP)ribose polymerase 3 (PARP3) as promoters of chromosomal rearrangements across human cell types. We focus on PARP3 as it is dispensable for murine viability and has druggable catalytic activity. We find that PARP3 regulates G quadruplex (G4) DNA in response to DNA damage, which suppresses repair by nonhomologous end-joining and homologous recombination. Chemical stabilization of G4 DNA in PARP3−/− cells leads to widespread DNA double-strand breaks and synthetic lethality. We propose a model in which PARP3 suppresses G4 DNA and facilitates DNA repair by multiple pathways., Chromosomal rearrangements are key events in the pathogenesis of a range of disorders. Here the authors utilize a zinc finger nuclease translocation reporter to identify PARP3 as a regulator of these events at sites enriched for G quadruplex DNA.
- Published
- 2017
10. Site-specific targeting of a light activated dCas9-KillerRed fusion protein generates transient, localized regions of oxidative DNA damage
- Author
-
Nealia C.M. House, Jacob V. Layer, Brendan D. Price, and Ramya Parasuram
- Subjects
DNA Repair ,Light ,Hydrolases ,Gene Expression ,Biochemistry ,Guide RNA ,chemistry.chemical_compound ,0302 clinical medicine ,CRISPR ,Clustered Regularly Interspaced Short Palindromic Repeats ,DNA Breaks, Double-Stranded ,0303 health sciences ,Ku70 ,Genome ,Multidisciplinary ,Chromosome Biology ,Chromatin ,Enzymes ,Cell biology ,Nucleic acids ,Non-homologous end joining ,030220 oncology & carcinogenesis ,293T cells ,Cell lines ,Medicine ,Epigenetics ,Biological cultures ,Research Article ,RNA, Guide, Kinetoplastida ,Nucleases ,DNA damage ,DNA repair ,Science ,Transfection ,Research and Analysis Methods ,Non-Homologous End Joining ,Cell Line ,03 medical and health sciences ,DNA-binding proteins ,Genetics ,Humans ,Molecular Biology Techniques ,Molecular Biology ,030304 developmental biology ,Biology and life sciences ,Cas9 ,Proteins ,DNA ,Cell Biology ,Endonucleases ,Oxidative Stress ,HEK293 Cells ,chemistry ,Enzymology ,RNA ,CRISPR-Cas Systems - Abstract
DNA repair requires reorganization of the local chromatin structure to facilitate access to and repair of the DNA. Studying DNA double-strand break (DSB) repair in specific chromatin domains has been aided by the use of sequence-specific endonucleases to generate targeted breaks. Here, we describe a new approach that combines KillerRed, a photosensitizer that generates reactive oxygen species (ROS) when exposed to light, and the genome-targeting properties of the CRISPR/Cas9 system. Fusing KillerRed to catalytically inactive Cas9 (dCas9) generates dCas9-KR, which can then be targeted to any desired genomic region with an appropriate guide RNA. Activation of dCas9-KR with green light generates a local increase in reactive oxygen species, resulting in “clustered” oxidative damage, including both DNA breaks and base damage. Activation of dCas9-KR rapidly (within minutes) increases both γH2AX and recruitment of the KU70/80 complex. Importantly, this damage is repaired within 10 minutes of termination of light exposure, indicating that the DNA damage generated by dCas9-KR is both rapid and transient. Further, repair is carried out exclusively through NHEJ, with no detectable contribution from HR-based mechanisms. Surprisingly, sequencing of repaired DNA damage regions did not reveal any increase in either mutations or INDELs in the targeted region, implying that NHEJ has high fidelity under the conditions of low level, limited damage. The dCas9-KR approach for creating targeted damage has significant advantages over the use of endonucleases, since the duration and intensity of DNA damage can be controlled in “real time” by controlling light exposure. In addition, unlike endonucleases that carry out multiple cut-repair cycles, dCas9-KR produces a single burst of damage, more closely resembling the type of damage experienced during acute exposure to reactive oxygen species or environmental toxins. dCas9-KR is a promising system to induce DNA damage and measure site-specific repair kinetics at clustered DNA lesions.
- Published
- 2020
11. HJURP knockdown disrupts clonogenic capacity and increasesradiation-induced cell death of glioblastoma cells
- Author
-
Christiane Pienna Soares, Brendan D. Price, Luis Fernando Macedo Di Cristofaro, Cibele Cardoso, Rodolfo Bortolozo Serafim, Wilson A. Silva, Enilza Maria Espreáfico, Valeria Valente, Maria Luísa Paço-Larson, Universidade de São Paulo (USP), Universidade Estadual Paulista (Unesp), Center for Cell-Based Therapy-CEPID/FAPESP, and Dana-Farber Cancer Institute
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,Cell cycle checkpoint ,Cell ,Biology ,Cell cycle ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Cell culture ,030220 oncology & carcinogenesis ,Radioresistance ,medicine ,Cancer research ,Molecular Medicine ,Gene silencing ,GLIOMA ,Clonogenic assay ,Molecular Biology - Abstract
Made available in DSpace on 2020-12-12T02:36:37Z (GMT). No. of bitstreams: 0 Previous issue date: 2020-05-01 Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) The Holliday Junction-Recognition Protein (HJURP) was reported as overexpressed in several cancers and also strongly correlated with poor prognosis of patients, especially in glioblastoma (GBM), the most common and deadly type of primary brain tumor. HJURP is responsible for loading the histone H3 variant—the Centromeric Protein A (CENP-A)—at the centromeres in a cell cycle-regulated manner, being required for proper chromosome segregation. Here we investigated HJURP association with survival and radioresistance of different GBM cell lines. HJURP knockdown compromised the clonogenic capacity and severely impaired survival of five distinct GBM cells, while nontumor astrocytes were not affected. U251MG cells showed a robust cell cycle arrest in G2/M phases followed by a drastic increment in cell death after HJURP silencing, while U138MG and U343MG cell lines presented augmented senescence with a comparable increase in cell death. Importantly, we verified that the impact on cell cycle dynamics and clonogenic survival were associated with loss CENP-A at the centromeres. Moreover, radiation resistance was also impacted by HJURP modulation in several GBM cell lines. U87MG, T98G, U138MG, and U343MG cells were all sensitized to ionizing radiation after HJURP reduction. These data reinforce the requirement of HJURP for proliferative capacity and radioresistance of tumor cells, underlining its potential as a promising therapeutic target for GBM. Department of Cellular and Molecular Biology Ribeirão Preto Medical School University of São Paulo (USP), Avenida Bandeirantes, 3900 São Paulo State University (UNESP) School of Pharmaceutical Sciences, Rodovia Araraquara - Jaú, Km 01 - s/n, Campos Ville Department of Genetics Ribeirão Preto Medical School University of São Paulo (USP), Avenida Bandeirantes, 3900 Center for Cell-Based Therapy-CEPID/FAPESP, Rua Tenente Catão Roxo, 2501 Center for Medical Genomics HCFMRP/USP and Center for Integrative System Biology - CISBi NAP/USP University of São Paulo Department of Radiation Oncology Dana-Farber Cancer Institute São Paulo State University (UNESP) School of Pharmaceutical Sciences, Rodovia Araraquara - Jaú, Km 01 - s/n, Campos Ville
- Published
- 2020
12. PDTM-41. INHIBITION OF DNA DOUBLE STRAND BREAK REPAIR PATHWAYS AS A PROMISING THERAPEUTIC TARGET IN DIFFUSE INTRINSIC PONTINE GLIOMAS (DIPGs)
- Author
-
Brendan D. Price, Dipanjan Chowdhury, Daphne A. Haas-Kogan, Jie Bian, and Sharmistha Pal
- Subjects
Genome instability ,Cancer Research ,DNA repair ,Chemistry ,DNA damage ,Pediatric Tumors ,Double-Stranded DNA Breaks ,Double Strand Break Repair ,chemistry.chemical_compound ,Oncology ,Cancer research ,Neurology (clinical) ,Personal Integrity ,Cell survival ,DNA - Abstract
New approaches to the treatment of diffuse intrinsic pontine gliomas (DIPGs) are desperately needed. DNA damage response is essential for cells to maintain genome integrity as DNA is damaged by both endogenous and exogenous stressors. Many cancer cells exhibit hyper-dependency on specific DNA repair pathways due to either defects in DNA repair mechanisms and/or high levels of endogenous stress leading to accumulation of DNA damage lesions. Identification of DIPG-specific DNA repair deficiencies and resultant dependencies may establish novel therapeutic strategies for DIPGs. METHODS To identify pathways critical for DIPG cell survival, genome wide CRISPR-Cas9 screen was performed on patient derived DIPG cell lines followed by gene set enrichment analyses. To monitor the effects of pathway inhibition on survival, apoptosis, DNA damage and repair, assays were performed to measure cell proliferation, cleaved-caspase3, gamma-H2AX and reporter based-DNA repair efficiency. RESULTS Our unbiased CRISPR approach to uncover vulnerabilities in DIPGs identified DNA double strand break (DSBs) repair pathways as essential for DIPG cell proliferation and survival. Further studies revealed high basal DSBs in DIPG cells compared to neural stem cells and primary astrocytes that suggest dependence of DIPG cell survival on specific DSB repair pathways. We confirmed the intrinsic reliance of DIPG cells on the specific DSB repair pathway of mutagenic end-joining, and defined a key role for DNA repair in suppressing endogenous DNA damage-induced apoptotic cell death. CONCLUSION DIPG cells have high endogenous DNA damage levels and escape catastrophic genomic instability and cell death by engaging DNA repair pathways, in particular the mutagenic end-joining DNA repair pathway. Inhibition of this specific DNA repair pathway represents a promising new avenue for the treatment of DIPGs.
- Published
- 2019
13. HJURP knockdown disrupts clonogenic capacity and increases radiation-induced cell death of glioblastoma cells
- Author
-
Rodolfo B, Serafim, Cibele, Cardoso, Luis F M, Di Cristofaro, Christiane, Pienna Soares, Wilson, Araújo Silva, Enilza M, Espreafico, Maria L, Paçó-Larson, Brendan D, Price, and Valeria, Valente
- Subjects
Brain Neoplasms ,Cell Survival ,Centromere ,Cell Cycle Checkpoints ,Radiation Tolerance ,DNA-Binding Proteins ,Cell Line, Tumor ,Gene Knockdown Techniques ,Neoplastic Stem Cells ,Humans ,Glioblastoma ,Centromere Protein A ,Tumor Stem Cell Assay ,Cell Proliferation - Abstract
The Holliday Junction-Recognition Protein (HJURP) was reported as overexpressed in several cancers and also strongly correlated with poor prognosis of patients, especially in glioblastoma (GBM), the most common and deadly type of primary brain tumor. HJURP is responsible for loading the histone H3 variant-the Centromeric Protein A (CENP-A)-at the centromeres in a cell cycle-regulated manner, being required for proper chromosome segregation. Here we investigated HJURP association with survival and radioresistance of different GBM cell lines. HJURP knockdown compromised the clonogenic capacity and severely impaired survival of five distinct GBM cells, while nontumor astrocytes were not affected. U251MG cells showed a robust cell cycle arrest in G2/M phases followed by a drastic increment in cell death after HJURP silencing, while U138MG and U343MG cell lines presented augmented senescence with a comparable increase in cell death. Importantly, we verified that the impact on cell cycle dynamics and clonogenic survival were associated with loss CENP-A at the centromeres. Moreover, radiation resistance was also impacted by HJURP modulation in several GBM cell lines. U87MG, T98G, U138MG, and U343MG cells were all sensitized to ionizing radiation after HJURP reduction. These data reinforce the requirement of HJURP for proliferative capacity and radioresistance of tumor cells, underlining its potential as a promising therapeutic target for GBM.
- Published
- 2019
14. Epigenetic therapy with inhibitors of histone methylation suppresses DNA damage signaling and increases glioma cell radiosensitivity
- Author
-
Brendan D. Price, Rodolfo Bortolozo Serafim, Chelsea Carman, Ozge Gursoy-Yuzugullu, Marios Myronakis, and Valeria Valente
- Subjects
0301 basic medicine ,Radiation-Sensitizing Agents ,Methyltransferase ,G9a ,DNA repair ,Biology ,Methylation ,Histones ,03 medical and health sciences ,0302 clinical medicine ,glioma ,Histone methylation ,Humans ,Cancer epigenetics ,Epigenetics ,histone methylation ,radiosensitizer ,EZH2 ,Molecular biology ,3. Good health ,030104 developmental biology ,Histone ,Oncology ,030220 oncology & carcinogenesis ,Histone methyltransferase ,Cancer research ,biology.protein ,Research Paper ,DNA Damage ,Signal Transduction - Abstract
// Ozge Gursoy-Yuzugullu 1 , Chelsea Carman 1 , Rodolfo Bortolozo Serafim 1 , Marios Myronakis 1 , Valeria Valente 2 , Brendan D. Price 1 1 Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA 02215, USA 2 Sao Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, Rodovia Araraquara-Jau, Campos Ville, SP, 14800-903, Brazil Correspondence to: Brendan D. Price, email: brendan_price@dfci.harvard.edu Keywords: DNA repair, radiosensitizer, glioma, histone methylation, G9a Received: January 04, 2017 Accepted: February 07, 2017 Published: February 20, 2017 ABSTRACT Radiation therapy is widely used to treat human malignancies, but many tumor types, including gliomas, exhibit significant radioresistance. Radiation therapy creates DNA double-strand breaks (DSBs), and DSB repair is linked to rapid changes in epigenetic modifications, including increased histone methylation. This increased histone methylation recruits DNA repair proteins which can then alter the local chromatin structure and promote repair. Consequently, combining inhibitors of specific histone methyltransferases with radiation therapy may increase tumor radiosensitivity, particularly in tumors with significant therapeutic resistance. Here, we demonstrate that inhibitors of the H4K20 methyltransferase SETD8 (UNC-0379) and the H3K9 methyltransferase G9a (BIX-01294) are effective radiosensitizers of human glioma cells. UNC-0379 blocked H4K20 methylation and reduced recruitment of the 53BP1 protein to DSBs, although this loss of 53BP1 caused only limited changes in radiosensitivity. In contrast, loss of H3K9 methylation through G9a inhibition with BIX-01294 increased radiosensitivity of a panel of glioma cells (SER2Gy range: 1.5 - 2.9). Further, loss of H3K9 methylation reduced DSB signaling dependent on H3K9, including reduced activation of the Tip60 acetyltransferase, loss of ATM signaling and reduced phosphorylation of the KAP-1 repressor. In addition, BIX-0194 inhibited DSB repair through both the homologous recombination and nonhomologous end-joining pathways. Inhibition of G9a and loss of H3K9 methylation is therefore an effective approach for increasing radiosensitivity of glioma cells. These results suggest that combining inhibitors of histone methyltransferases which are critical for DSB repair with radiation therapy may provide a new therapeutic route for sensitizing gliomas and other tumors to radiation therapy.
- Published
- 2017
15. Histone chaperone Anp32e removes H2A.Z from DNA double-strand breaks and promotes nucleosome reorganization and DNA repair
- Author
-
Marina K. Ayrapetov, Brendan D. Price, and Ozge Gursoy-Yuzugullu
- Subjects
animal structures ,Multidisciplinary ,Biological Sciences ,Biology ,Linker DNA ,Molecular biology ,Cell biology ,Histone H4 ,Histone ,embryonic structures ,Histone methylation ,Histone H2A ,biology.protein ,Nucleosome ,Histone code ,Histone octamer - Abstract
The repair of DNA double-strand breaks (DSBs) requires open, flexible chromatin domains. The NuA4–Tip60 complex creates these flexible chromatin structures by exchanging histone H2A.Z onto nucleosomes and promoting acetylation of histone H4. Here, we demonstrate that the accumulation of H2A.Z on nucleosomes at DSBs is transient, and that rapid eviction of H2A.Z is required for DSB repair. Anp32e, an H2A.Z chaperone that interacts with the C-terminal docking domain of H2A.Z, is rapidly recruited to DSBs. Anp32e functions to remove H2A.Z from nucleosomes, so that H2A.Z levels return to basal within 10 min of DNA damage. Further, H2A.Z removal by Anp32e disrupts inhibitory interactions between the histone H4 tail and the nucleosome surface, facilitating increased acetylation of histone H4 following DNA damage. When H2A.Z removal by Anp32e is blocked, nucleosomes at DSBs retain elevated levels of H2A.Z, and assume a more stable, hypoacetylated conformation. Further, loss of Anp32e leads to increased CtIP-dependent end resection, accumulation of single-stranded DNA, and an increase in repair by the alternative nonhomologous end joining pathway. Exchange of H2A.Z onto the chromatin and subsequent rapid removal by Anp32e are therefore critical for creating open, acetylated nucleosome structures and for controlling end resection by CtIP. Dynamic modulation of H2A.Z exchange and removal by Anp32e reveals the importance of the nucleosome surface and nucleosome dynamics in processing the damaged chromatin template during DSB repair.
- Published
- 2015
- Full Text
- View/download PDF
16. Ape1 guides DNA repair pathway choice that is associated with drug tolerance in glioblastoma
- Author
-
Thomas Ströbel, Adelheid Wöhrer, Sarah Vose, Klemens Vierlinger, Sibylle Madlener, Brendan D. Price, Tonny Lagerweij, Thomas Wurdinger, Serkan Tuna, Okay Saydam, Bruce Demple, Nurten Saydam, Neurosurgery, and CCA - Cancer Treatment and quality of life
- Subjects
0301 basic medicine ,DNA Repair ,DNA repair ,Ubiquitin-Protein Ligases ,RAD51 ,lcsh:Medicine ,Article ,03 medical and health sciences ,Endonuclease ,chemistry.chemical_compound ,0302 clinical medicine ,DNA-(Apurinic or Apyrimidinic Site) Lyase ,Humans ,AP site ,Homologous Recombination ,lcsh:Science ,Multidisciplinary ,biology ,Kinase ,lcsh:R ,Drug Tolerance ,DNA Repair Pathway ,Molecular biology ,Checkpoint Kinase 2 ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,lcsh:Q ,Glioblastoma ,Homologous recombination ,Metabolic Networks and Pathways ,DNA - Abstract
Ape1 is the major apurinic/apyrimidinic (AP) endonuclease activity in mammalian cells, and a key factor in base-excision repair of DNA. High expression or aberrant subcellular distribution of Ape1 has been detected in many cancer types, correlated with drug response, tumor prognosis, or patient survival. Here we present evidence that Ape1 facilitates BRCA1-mediated homologous recombination repair (HR), while counteracting error-prone non-homologous end joining of DNA double-strand breaks. Furthermore, Ape1, coordinated with checkpoint kinase Chk2, regulates drug response of glioblastoma cells. Suppression of Ape1/Chk2 signaling in glioblastoma cells facilitates alternative means of damage site recruitment of HR proteins as part of a genomic defense system. Through targeting “HR-addicted” temozolomide-resistant glioblastoma cells via a chemical inhibitor of Rad51, we demonstrated that targeting HR is a promising strategy for glioblastoma therapy. Our study uncovers a critical role for Ape1 in DNA repair pathway choice, and provides a mechanistic understanding of DNA repair-supported chemoresistance in glioblastoma cells.
- Published
- 2017
- Full Text
- View/download PDF
17. Human CHD1 is required for early DNA-damage signaling and is uniquely regulated by its N terminus
- Author
-
Rodolfo Bortolozo Serafim, Catarina G. Ferreira, Timur Yusufzai, Brendan D. Price, Jiaqi Li, Kathryn A. Coe, Steven Ketchum, Jia Zhou, and Jessica Liu
- Subjects
0301 basic medicine ,DNA damage ,Poly ADP ribose polymerase ,Biology ,Chromatin remodeling ,Cell Line ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,Gene Knockout Techniques ,0302 clinical medicine ,Genetics ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,Homologous Recombination ,Gene ,Adenosine Triphosphatases ,Binding Sites ,Endodeoxyribonucleases ,Gene regulation, Chromatin and Epigenetics ,DNA Helicases ,Nuclear Proteins ,Chromatin Assembly and Disassembly ,Peptide Fragments ,Recombinant Proteins ,Cell biology ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Homologous recombination ,Carrier Proteins ,Chromatin immunoprecipitation ,DNA ,DNA Damage ,Signal Transduction - Abstract
CHD1 is a conserved chromatin remodeling enzyme required for development and linked to prostate cancer in adults, yet its role in human cells is poorly understood. Here, we show that targeted disruption of the CHD1 gene in human cells leads to a defect in early double-strand break (DSB) repair via homologous recombination (HR), resulting in hypersensitivity to ionizing radiation as well as PARP and PTEN inhibition. CHD1 knockout cells show reduced H2AX phosphorylation (γH2AX) and foci formation as well as impairments in CtIP recruitment to the damaged sites. Chromatin immunoprecipitation following a single DSB shows that the reduced levels of γH2AX accumulation at DSBs in CHD1-KO cells are due to both a global reduction in H2AX incorporation and poor retention of H2AX at the DSBs. We also identified a unique N-terminal region of CHD1 that inhibits the DNA binding, ATPase, and chromatin assembly and remodeling activities of CHD1. CHD1 lacking the N terminus was more active in rescuing the defects in γH2AX formation and CtIP recruitment in CHD1-KO cells than full-length CHD1, suggesting the N terminus is a negative regulator in cells. Our data point to a role for CHD1 in the DSB repair process and identify a novel regulatory region of the protein.
- Published
- 2017
18. Spatially restricted loading of BRD2 at DNA double-strand breaks protects H4 acetylation domains and promotes DNA repair
- Author
-
Chelsea Carman, Brendan D. Price, and Ozge Gursoy-Yuzugullu
- Subjects
0301 basic medicine ,DNA End-Joining Repair ,DNA Repair ,HMG-box ,Chromosomal Proteins, Non-Histone ,genetic processes ,lcsh:Medicine ,Protein Serine-Threonine Kinases ,Article ,Chromatin remodeling ,Histones ,Histone H4 ,03 medical and health sciences ,Valosin Containing Protein ,Cell Line, Tumor ,Humans ,DNA Breaks, Double-Stranded ,lcsh:Science ,KAT5 ,ChIA-PET ,Multidisciplinary ,biology ,Tumor Suppressor Proteins ,lcsh:R ,fungi ,Acetylation ,Molecular biology ,Bromodomain ,Chromatin ,Cell biology ,Repressor Proteins ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Histone ,health occupations ,biology.protein ,lcsh:Q ,biological phenomena, cell phenomena, and immunity ,Tumor Suppressor p53-Binding Protein 1 ,Protein Binding ,Transcription Factors - Abstract
The n-terminal tail of histone H4 recruits repair proteins, including 53BP1, to DNA double-strand breaks (DSB) and undergoes dynamic acetylation during DSB repair. However, how H4 acetylation (H4Ac) recruits repair proteins and reorganizes chromatin during DNA repair is unclear. Here, we show that the bromodomain protein BRD2 is recruited to DSBs. This recruitment requires binding of BRD2’s tandem bromodomains to H4Ac, which is generated at DSBs by the Tip60/KAT5 acetyltransferase. Binding of BRD2 to H4Ac protects the underlying acetylated chromatin from attack by histone deacetylases and allows acetylation to spread along the flanking chromatin. However, BRD2 recruitment is spatially restricted to a chromatin domain extending only 2 kb either side of the DSB, and BRD2 does not spread into the chromatin domains flanking the break. Instead, BRD2 facilitates recruitment of a second bromodomain protein, ZMYND8, which spreads along the flanking chromatin, but is excluded from the DSB region. This creates a spatially restricted H4Ac/BRD2 domain which reorganizes chromatin at DSBs, limits binding of the L3MBTL1 repressor and promotes 53BP1 binding, while limiting end-resection of DSBs. BRD2 therefore creates a restricted chromatin environment surrounding DSBs which facilitates DSB repair and which is framed by extensive ZMYND8 domains on the flanking chromatin.
- Published
- 2017
- Full Text
- View/download PDF
19. KDM5A demethylase: Erasing histone modifications to promote repair of DNA breaks
- Author
-
Brendan D. Price
- Subjects
0301 basic medicine ,DNA Repair ,DNA repair ,Cell ,behavioral disciplines and activities ,environment and public health ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Histone H2A ,medicine ,Histone code ,Spotlight ,biology ,DNA Breaks ,Cell Biology ,Molecular biology ,Chromatin ,Histone Code ,030104 developmental biology ,Histone ,medicine.anatomical_structure ,KDM5A ,biology.protein ,Commentary ,Demethylase ,030217 neurology & neurosurgery - Abstract
Price previews work from the Miller laboratory identifying the H3K4 histone demethylase KDM5A as promoting double-stranded DNA break repair., Repairing DNA breaks within the complexity of the cell chromatin is challenging. In this issue, Gong et al. (2017. J. Cell Biol. https://doi.org/10.1083/jcb.201611135) identify the histone demethylase KDM5A as a critical editor of the cells’ “histone code” that is required to recruit DNA repair complexes to DNA breaks.
- Published
- 2017
20. Essential role for mammalian apurinic/apyrimidinic (AP) endonuclease Ape1/Ref-1 in telomere maintenance
- Author
-
Bruce Demple, Thomas Ströbel, Sarah Vose, Okay Saydam, Nurten Saydam, Brendan D. Price, and Sibylle Madlener
- Subjects
Telomere-binding protein ,Chromatin Immunoprecipitation ,Telomerase ,Multidisciplinary ,DNA Repair ,biology ,DNA repair ,Blotting, Western ,Fluorescent Antibody Technique ,Telomere Homeostasis ,Biological Sciences ,DNA-(apurinic or apyrimidinic site) lyase ,Molecular biology ,AP endonuclease ,Telomere ,Cell Line, Tumor ,DNA-(Apurinic or Apyrimidinic Site) Lyase ,biology.protein ,Humans ,Immunoprecipitation ,AP site ,In Situ Hybridization, Fluorescence ,DNA Primers - Abstract
The major mammalian apurinic/apyrimidinic endonuclease Ape1 is a multifunctional protein operating in protection of cells from oxidative stress via its DNA repair, redox, and transcription regulatory activities. The importance of Ape1 has been marked by previous work demonstrating its requirement for viability in mammalian cells. However, beyond a requirement for Ape1-dependent DNA repair activity, deeper molecular mechanisms of the fundamental role of Ape1 in cell survival have not been defined. Here, we report that Ape1 is an essential factor stabilizing telomeric DNA, and its deficiency is associated with telomere dysfunction and segregation defects in immortalized cells maintaining telomeres by either the alternative lengthening of telomeres pathway (U2OS) or telomerase expression (BJ-hTERT), or in normal human fibroblasts (IMR90). Through the expression of Ape1 derivatives with site-specific changes, we found that the DNA repair and N-terminal acetylation domains are required for the Ape1 function at telomeres. Ape1 associates with telomere proteins in U2OS cells, and Ape1 depletion causes dissociation of TRF2 protein from telomeres. Consistent with this effect, we also observed that Ape1 depletion caused telomere shortening in both BJ-hTERT and in HeLa cells. Thus, our study describes a unique and unpredicted role for Ape1 in telomere protection, providing a direct link between base excision DNA repair activities and telomere metabolism.
- Published
- 2013
- Full Text
- View/download PDF
21. FANCD2 Activates Transcription of TAp63 and Suppresses Tumorigenesis
- Author
-
Alea A. Mills, Jung Min Kim, Alan D. D'Andrea, Benjamin Primack, Brendan D. Price, Eunmi Park, Sofia Vidal-Cardenas, Hyungjin Kim, and Ye Xu
- Subjects
Skin Neoplasms ,Cell ,medicine.disease_cause ,Mice ,0302 clinical medicine ,Fanconi anemia ,hemic and lymphatic diseases ,Monoubiquitination ,Genes, Tumor Suppressor ,Promoter Regions, Genetic ,Cells, Cultured ,Cellular Senescence ,Disease Resistance ,Mice, Knockout ,0303 health sciences ,education.field_of_study ,Fanconi Anemia Complementation Group D2 Protein ,3. Good health ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Female ,Ubiquitin-Specific Proteases ,Cell aging ,Protein Binding ,Transcriptional Activation ,congenital, hereditary, and neonatal diseases and abnormalities ,Population ,Biology ,Article ,03 medical and health sciences ,FANCD2 ,Endopeptidases ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Neoplasms, Squamous Cell ,education ,Molecular Biology ,030304 developmental biology ,Cell Proliferation ,Cell growth ,Arabidopsis Proteins ,Ubiquitination ,nutritional and metabolic diseases ,Cell Biology ,medicine.disease ,Phosphoproteins ,Molecular biology ,Mice, Inbred C57BL ,Fanconi Anemia ,Genes, ras ,Cancer research ,Trans-Activators ,Carcinogenesis ,DNA Damage - Abstract
Fanconi anemia (FA) is a rare genetic disorder characterized by an increased susceptibility to squamous cell cancers. Fifteen FA genes are known, and the encoded proteins cooperate in a common DNA repair pathway. A critical step is the monoubiquitination of the FANCD2 protein, and cells from most FA patients are deficient in this step. How monoubiquitinated FANCD2 suppresses squamous cell cancers is unknown. Here we show that Fancd2-deficient mice are prone to Ras-oncogene-driven skin carcinogenesis, while Usp1-deficient mice, expressing elevated cellular levels of Fancd2-Ub, are resistant to skin tumors. Moreover, Fancd2-Ub activates the transcription of the tumor suppressor TAp63, thereby promoting cellular senescence and blocking skin tumorigenesis. For FA patients, the reduction of FANCD2-Ub and TAp63 protein levels may account for their susceptibility to squamous cell neoplasia. Taken together, Usp1 inhibition may be a useful strategy for upregulating TAp63 and preventing or treating squamous cell cancers in the general non-FA population.
- Published
- 2013
- Full Text
- View/download PDF
22. Chromatin Remodeling at DNA Double-Strand Breaks
- Author
-
Alan D. D'Andrea and Brendan D. Price
- Subjects
Genome instability ,DNA Repair ,DNA repair ,genetic processes ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,Genomic Instability ,Article ,Histones ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,Nucleosome ,Animals ,Humans ,DNA Breaks, Double-Stranded ,030304 developmental biology ,Double strand ,0303 health sciences ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,fungi ,Chromatin Assembly and Disassembly ,Molecular biology ,Chromatin ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Histone ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,health occupations ,biological phenomena, cell phenomena, and immunity ,DNA - Abstract
DNA double-strand breaks (DSBs) can arise from multiple sources, including exposure to ionizing radiation. The repair of DSBs involves both post-translational modification of nucleosomes and concentration of DNA repair proteins at the site of damage. Consequently, nucleosome packing and chromatin architecture surrounding the DSB may limit the ability of the DNA damage response to access and repair the break. Here, we review early chromatin-based events that promote the formation of open, relaxed chromatin structures at DSBs and which allow the DNA repair machinery to access the spatially confined region surrounding the DSB, thereby facilitating mammalian DSB repair.
- Published
- 2013
- Full Text
- View/download PDF
23. DNA Damage Enhancement from Gold Nanoparticles for Clinical MV Photon Beams
- Author
-
Wilfred Ngwa, Janki Patel, G. Mike Makrigiorgos, Rajiv Kumar, Brendan D. Price, Alec C. Kimmelman, Houari Korideck, Sarah Johnson, Srinivas Sridhar, and Ross Berbeco
- Subjects
Photons ,Radiation ,Materials science ,Photon ,Aperture ,DNA damage ,Biophysics ,Metal Nanoparticles ,Isocenter ,Nanotechnology ,Delivery mode ,Article ,Colloidal gold ,Humans ,Radiology, Nuclear Medicine and imaging ,Gold ,Irradiation ,Beam (structure) ,DNA Damage ,HeLa Cells ,Biomedical engineering - Abstract
In this study, we quantify the relative damage enhancement due to the presence of gold nanoparticles (GNP) in vitro in a clinical 6 MV beam for various delivery parameters and depths. It is expected that depths and delivery modes that produce a larger proportions of low-energy photons will have a larger effect on the cell samples containing GNP. HeLa cells with and without 50 nm GNP were irradiated at depths of 1.5, 5, 10, 15 and 20 cm. Conventional beams with square aperture sizes 5, 10 and 15 cm at isocenter, and flattening filter free (FFF) beams were used. Relative DNA damage enhancement with GNP was evaluated by γ-H2AX staining. Statistically significant increases in DNA damage with GNP, compared to the absence of GNP, were observed for all depths and delivery modes. Relative to the shallowest depth, damage enhancement was observed to increase as a function of increasing depth for all deliveries. For the conventional (open field) delivery, DNA damage enhancement with GNP was seen to increase as a function of field size. For FFF delivery, a substantial increase in enhancement was found relative to the conventional field delivery. The measured relative DNA damage enhancement validates the theoretically predicted trends as a function of depth and delivery mode for clinical MV photon beams. The results of this study open new possibilities for the clinical development of gold nanoparticle-aided radiation therapy.
- Published
- 2012
- Full Text
- View/download PDF
24. Histone H2A.Z Controls a Critical Chromatin Remodeling Step Required for DNA Double-Strand Break Repair
- Author
-
Marina K. Ayrapetov, Yiduo Hu, Brendan D. Price, Ozge Gursoy-Yuzugullu, Chang Xu, and Ye Xu
- Subjects
Histone-modifying enzymes ,DNA End-Joining Repair ,Time Factors ,DNA Repair ,genetic processes ,Solenoid (DNA) ,Transfection ,Binding, Competitive ,Chromatin remodeling ,Article ,Histones ,Histone H1 ,Nucleosome ,Histone code ,Humans ,DNA Breaks, Double-Stranded ,Ku Autoantigen ,Molecular Biology ,Adenosine Triphosphatases ,Binding Sites ,Endodeoxyribonucleases ,biology ,fungi ,Ubiquitination ,Nuclear Proteins ,Acetylation ,Antigens, Nuclear ,Dose-Response Relationship, Radiation ,Cell Biology ,Chromatin Assembly and Disassembly ,Cell biology ,Chromatin ,Nucleosomes ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,Histone ,HEK293 Cells ,Biochemistry ,biology.protein ,Nucleic Acid Conformation ,RNA Interference ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins ,HeLa Cells - Abstract
Chromatin remodeling during DNA double-strand break (DSB) repair is required to facilitate access to and repair of DSBs. This remodeling requires increased acetylation of histones and a shift in nucleosome organization to create open, relaxed chromatin domains. However, the underlying mechanism driving changes in nucleosome structure at DSBs is poorly defined. Here, we demonstrate that histone H2A.Z is exchanged onto nucleosomes at DSBs by the p400 remodeling ATPase. H2A.Z exchange at DSBs shifts the chromatin to an open conformation and is required for acetylation and ubiquitination of histones and for loading of the brca1 complex. H2A.Z exchange also restricts single-stranded DNA production by nucleases and is required for loading of the Ku70/Ku80 DSB repair protein. H2A.Z exchange therefore promotes specific patterns of histone modification and reorganization of the chromatin architecture, leading to the assembly of a chromatin template that is an efficient substrate for the DSB repair machinery.
- Published
- 2012
- Full Text
- View/download PDF
25. Chromatin dynamics and the repair of DNA double strand breaks
- Author
-
Brendan D. Price and Ye Xu
- Subjects
Histone-modifying enzymes ,DNA Repair ,cells ,genetic processes ,Review ,Solenoid (DNA) ,Biology ,Chromatin remodeling ,Histones ,Nucleosome ,Histone code ,DNA Breaks, Double-Stranded ,Chromatin structure remodeling (RSC) complex ,Molecular Biology ,fungi ,Ubiquitination ,Cell Biology ,Molecular biology ,Chromatin ,Nucleosomes ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Histone ,health occupations ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Developmental Biology - Abstract
DNA double-strand breaks (DSBs) arise through both replication errors and from exogenous events such as exposure to ionizing radiation. DSBs are potentially lethal, and cells have evolved a highly conserved mechanism to detect and repair these lesions. This mechanism involves phosphorylation of histone H2AX (γH2AX) and the loading of DNA repair proteins onto the chromatin adjacent to the DSB. It is now clear that the chromatin architecture in the region surrounding the DSB has a critical impact on the ability of cells to mount an effective DNA damage response. DSBs promote the direct the formation of open, relaxed chromatin domains which are spatially confined to the area surrounding the break. These relaxed chromatin structures are created through the coupled action of the p400 SWI/SNF ATPase and histone acetylation by the Tip60 acetyltransferase. The resulting destabilization of nucleosomes at the DSB by Tip60 and p400 is required for ubiquitination of the chromatin by the RNF8 ubiquitin ligase, and for the subsequent recruitment of the brca1 complex. Chromatin dynamics at DSBs can therefore exert a powerful influence on the process of DSB repair. Further, there is emerging evidence that the different chromatin structures in the cell, such as heterochromatin and euchromatin, utilize distinct remodeling complexes and pathways to facilitate DSB. The processing and repair of DSB is therefore critically influenced by the nuclear architecture in which the lesion arises.
- Published
- 2011
- Full Text
- View/download PDF
26. The p400 ATPase regulates nucleosome stability and chromatin ubiquitination during DNA repair
- Author
-
Shenghong Yang, Yingli Sun, David M. Weinstock, Xiaofeng Jiang, Marina K. Ayrapetov, Brendan D. Price, Patryk Moskwa, and Ye Xu
- Subjects
endocrine system diseases ,DNA Repair ,cells ,Solenoid (DNA) ,Biology ,Lysine Acetyltransferase 5 ,Article ,Chromatin remodeling ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Nucleosome ,Histone code ,Chromatin structure remodeling (RSC) complex ,skin and connective tissue diseases ,Research Articles ,Histone Acetyltransferases ,030304 developmental biology ,0303 health sciences ,Protein Stability ,DNA Helicases ,Ubiquitination ,Cell Biology ,Chromatin Assembly and Disassembly ,Molecular biology ,Chromatin ,SWI/SNF ,Nucleosomes ,3. Good health ,Cell biology ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,Histone ,030220 oncology & carcinogenesis ,biology.protein ,biological phenomena, cell phenomena, and immunity ,DNA Damage - Abstract
p400 unwinds chromatin from nucleosomes flanking double-strand breaks to facilitate recruitment of the DNA repair components brca1 and 53BP1., The complexity of chromatin architecture presents a significant barrier to the ability of the DNA repair machinery to access and repair DNA double-strand breaks (DSBs). Consequently, remodeling of the chromatin landscape adjacent to DSBs is vital for efficient DNA repair. Here, we demonstrate that DNA damage destabilizes nucleosomes within chromatin regions that correspond to the γ-H2AX domains surrounding DSBs. This nucleosome destabilization is an active process requiring the ATPase activity of the p400 SWI/SNF ATPase and histone acetylation by the Tip60 acetyltransferase. p400 is recruited to DSBs by a mechanism that is independent of ATM but requires mdc1. Further, the destabilization of nucleosomes by p400 is required for the RNF8-dependent ubiquitination of chromatin, and for the subsequent recruitment of brca1 and 53BP1 to DSBs. These results identify p400 as a novel DNA damage response protein and demonstrate that p400-mediated alterations in nucleosome and chromatin structure promote both chromatin ubiquitination and the accumulation of brca1 and 53BP1 at sites of DNA damage.
- Published
- 2010
- Full Text
- View/download PDF
27. Autophagy: A new target for advanced papillary thyroid cancer therapy
- Author
-
Francis D. Moore, Brendan D. Price, Edward E. Whang, Michael A. Abramson, Daniel T. Ruan, David B. Donner, Xiaofeng Jiang, and Chi-Iou Lin
- Subjects
Programmed cell death ,medicine.medical_specialty ,endocrine system diseases ,In Vitro Techniques ,Radiation Tolerance ,Papillary thyroid cancer ,Cell Line, Tumor ,Internal medicine ,Autophagy ,medicine ,Humans ,Doxorubicin ,Thyroid Neoplasms ,Radiosensitivity ,Thyroid cancer ,Antibiotics, Antineoplastic ,business.industry ,Adenine ,Cancer ,medicine.disease ,Carcinoma, Papillary ,Endocrinology ,Drug Resistance, Neoplasm ,Cell culture ,Cancer research ,Surgery ,business ,Microtubule-Associated Proteins ,medicine.drug - Abstract
Background Autophagy is a conserved response to stress that facilitates cell survival in some contexts and promotes cell death in others. We sought to characterize autophagy in papillary thyroid cancer (PTC), and to determine the effects of autophagy inhibition on chemosensitivity and radiosensitivity. Methods The human thyroid papillary carcinoma cell lines TPC-1 and 8505-C were treated with doxorubicin or radiation in the presence or absence of the autophagy-specific inhibitor 3-methyladenine (3-MA). Results Although light chain 3 (LC3)-II protein levels were undetectable in normal thyroid and PTC specimens at baseline, doxorubicin exposure induced LC3-II expression and the formation of autophagosomes. Both PTC cell lines expressed low levels of LC3-II under standard conditions. Treatment of these cells with doxorubicin strongly induced LC3-II expression and the formation of autophagosomes; however, doxorubicin–mediated induction of LC3-II was abrogated by 3-MA. Moreover, 3-MA significantly increased the doxorubicin IC 50 in both PTC cell lines. Radiation exposure also induced LC3-II expression. Treatment with 3-MA abrogated the radiation–induced increase in LC3-II in both cell lines and reduced radiosensitivity by 49% and 31% in 8505-C and TPC-1 cells, respectively. Conclusion Autophagy inhibition promotes PTC resistance to doxorubicin and radiation. Therefore, autophagy activation may be a useful adjunct treatment for patients with PTC that is refractory to conventional therapy.
- Published
- 2009
- Full Text
- View/download PDF
28. Galectin-3 Targeted Therapy with a Small Molecule Inhibitor Activates Apoptosis and Enhances Both Chemosensitivity and Radiosensitivity in Papillary Thyroid Cancer
- Author
-
Francis D. Moore, Edward E. Whang, Adelaide M. Carothers, Brendan D. Price, Vania Nosé, Xiaofeng Jiang, Ulf J. Nilsson, Daniel T. Ruan, David B. Donner, Chi-Iou Lin, Tamara Delaine, and Hakon Leffler
- Subjects
Radiation-Sensitizing Agents ,Cancer Research ,endocrine system diseases ,Galectin 3 ,medicine.medical_treatment ,Poly (ADP-Ribose) Polymerase-1 ,Apoptosis ,Caspase 3 ,Radiation Tolerance ,Thiogalactosides ,Papillary thyroid cancer ,Targeted therapy ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Doxorubicin ,Thyroid Neoplasms ,Radiosensitivity ,Clonogenic assay ,Molecular Biology ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemistry ,Drug Synergism ,Flow Cytometry ,medicine.disease ,Carcinoma, Papillary ,Rats ,Oncology ,Drug Resistance, Neoplasm ,Thioglycosides ,Cancer research ,Poly(ADP-ribose) Polymerases ,Ex vivo ,medicine.drug - Abstract
Although most patients with papillary thyroid cancer (PTC) have favorable outcomes, some have advanced PTC that is refractory to external beam radiation and systemic chemotherapy. Galectin-3 (Gal-3) is a β-galactoside–binding protein with antiapoptotic activity that is consistently overexpressed in PTC. The purpose of this study is to determine if Gal-3 inhibition promotes apoptosis, chemosensitivity, and radiosensitivity in PTC. PTC cell lines (8505-C and TPC-1) and human ex vivo PTC were treated with a highly specific small molecule inhibitor of Gal-3 (Td131_1). Apoptotic activity was determined by flow cytometric analysis as well as caspase-3 and PARP cleavage. The minimum inhibitory concentrations of Td131_1 and doxorubicin were determined, and their combined effects were measured to test for synergistic activity. The effects of Td131_1 on radiosensitivity were determined by a clonogenic assay. Td131_1 promoted apoptosis, improved radiosensitivity, and synergistically enhanced chemosensitivity to doxorubicin in PTC cell lines. In PTC ex vivo, Td131_1 treatment alone induced the cleavage of caspase-3 and PARP. Td131_1 and doxorubicin together activated apoptosis in PTC ex vivo to a greater degree than their combined individual effects. Td131_1 activated apoptosis and had synergistic activity with doxorubicin in PTC. We conclude that Gal-3 targeted therapy is a promising therapeutic strategy for advanced PTC that is refractory to surgery and radioactive iodine therapy. (Mol Cancer Res 2009;7(10):1655–62)
- Published
- 2009
- Full Text
- View/download PDF
29. Histone H3 methylation links DNA damage detection to activation of the Tip60 tumor suppressor
- Author
-
Johnathan R. Whetstine, Marina K. Ayrapetov, Lisa A. Moreau, Brendan D. Price, Yingli Sun, Ye Xu, and Xiaofeng Jiang
- Subjects
DNA repair ,Lysine Acetyltransferase 5 ,Article ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Histone methylation ,Histone H2A ,Humans ,Genes, Tumor Suppressor ,Cancer epigenetics ,KAT5 ,030304 developmental biology ,Histone Acetyltransferases ,0303 health sciences ,biology ,Cell Biology ,DNA Methylation ,Molecular biology ,3. Good health ,Cell biology ,Histone ,Chromobox Protein Homolog 5 ,030220 oncology & carcinogenesis ,Acetyltransferase ,biology.protein ,DNA Damage ,HeLa Cells - Abstract
DNA double-strand break (DSB) repair involves complex interactions between chromatin and repair proteins, including Tip60, a tumour suppressor. Tip60 is an acetyltransferase that acetylates both histones and ATM (ataxia telangiectasia mutated) kinase. Inactivation of Tip60 leads to defective DNA repair and increased cancer risk. However, how DNA damage activates the acetyltransferase activity of Tip60 is not known. Here, we show that direct interaction between the chromodomain of Tip60 and histone H3 trimethylated on lysine 9 (H3K9me3) at DSBs activates the acetyltransferase activity of Tip60. Depletion of intracellular H3K9me3 blocks activation of the acetyltransferase activity of Tip60, resulting in defective ATM activation and widespread defects in DSB repair. In addition, the ability of Tip60 to access H3K9me3 is dependent on the DNA damage-induced displacement of HP1beta (heterochromatin protein 1beta) from H3K9me3. Finally, we demonstrate that the Mre11-Rad50-Nbs1 (MRN) complex targets Tip60 to H3K9me3, and is required to activate the acetyltransferase activity of Tip60. These results reveal a new function for H3K9me3 in coordinating activation of Tip60-dependent DNA repair pathways, and imply that aberrant patterns of histone methylation may contribute to cancer by altering the efficiency of DSB repair.
- Published
- 2009
30. Patching broken DNA: Nucleosome dynamics and the repair of DNA breaks
- Author
-
Brendan D. Price, Ozge Gursoy-Yuzugullu, and Nealia C.M. House
- Subjects
0301 basic medicine ,biology ,DNA Repair ,Chromatin Assembly and Disassembly ,Molecular biology ,Mi-2/NuRD complex ,Models, Biological ,Chromatin remodeling ,Article ,Chromatin ,Cell biology ,Nucleosomes ,03 medical and health sciences ,030104 developmental biology ,Histone ,DNA Repair Enzymes ,Structural Biology ,Multienzyme Complexes ,Histone methylation ,Histone H2A ,biology.protein ,Nucleosome ,Histone code ,DNA Breaks, Double-Stranded ,Molecular Biology - Abstract
The ability of cells to detect and repair DNA double-strand breaks (DSBs) is dependent on reorganization of the surrounding chromatin structure by chromatin remodeling complexes. These complexes promote access to the site of DNA damage, facilitate processing of the damaged DNA and, importantly, are essential to repackage the repaired DNA. Here, we will review the chromatin remodeling steps which occur immediately after DSB production and which prepare the damaged chromatin template for processing by the DSB repair machinery. DSBs promote rapid accumulation of repressive complexes, including HP1, the NuRD complex, H2A.Z and histone methyltransferases at the DSB. This shift to a repressive chromatin organization may be important to inhibit local transcription and limit mobility of the break, and to maintain the DNA ends in close contact. Subsequently, the repressive chromatin is rapidly dismantled through a mechanism involving dynamic exchange of the histone variant H2A.Z. H2A.Z removal at DSBs alters the acidic patch on the nucleosome surface, promoting acetylation of the H4 tail (by the NuA4-Tip60 complex) and shifting the chromatin to a more open structure. Further, H2A.Z removal promotes chromatin ubiquitination and recruitment of additional DSB repair proteins to the break. Modulation of the nucleosome surface and nucleosome function during DSB repair therefore plays a vital role in processing of DNA breaks. Further, the nucleosome surface may function as a central hub during DSB repair, directing specific patterns of histone modification, recruiting DNA repair proteins and modulating chromatin packing during processing of the damaged DNA template.
- Published
- 2015
31. Inhibition of histone acetyltransferase activity by anacardic acid sensitizes tumor cells to ionizing radiation
- Author
-
Brendan D. Price, Xiaofeng Jiang, Shujuan Chen, and Yingli Sun
- Subjects
Radiation-Sensitizing Agents ,DNA repair ,DNA damage ,animal diseases ,Biophysics ,Anacardic acid ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,DNA-Activated Protein Kinase ,Protein Serine-Threonine Kinases ,environment and public health ,Biochemistry ,Lysine Acetyltransferase 5 ,Radiosensitivity ,chemistry.chemical_compound ,Structural Biology ,Cell Line, Tumor ,Neoplasms ,Radiation, Ionizing ,parasitic diseases ,Genetics ,Humans ,Histone acetyltransferase activity ,Enzyme Inhibitors ,Molecular Biology ,Histone Acetyltransferases ,biology ,Tumor Suppressor Proteins ,Cell Biology ,Histone acetyltransferase ,Molecular biology ,Anacardic Acids ,Cell biology ,Chromatin ,Anacardic acids ,DNA-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,Tip60 ,chemistry ,ATM ,HAT ,embryonic structures ,biology.protein ,Histone deacetylase ,HeLa Cells - Abstract
Histone acetyltransferases (HATs) regulate transcription, chromatin structure and DNA repair. Here, we utilized a novel HAT inhibitor, anacardic acid, to examine the role of HATs in the DNA damage response. Anacardic acid inhibits the Tip60 HAT in vitro, and blocks the Tip60-dependent activation of the ATM and DNA–PKcs protein kinases by DNA damage in vivo. Further, anacardic acid sensitizes human tumor cells to the cytotoxic effects of ionizing radiation. These results demonstrate a central role for HATs such as Tip60 in regulating the DNA damage response. HAT inhibitors provide a novel therapeutic approach for increasing the sensitivity of tumors to radiation therapy.
- Published
- 2006
- Full Text
- View/download PDF
32. A role for the Tip60 histone acetyltransferase in the acetylation and activation of ATM
- Author
-
Norvin Fernandes, Shujuan Chen, Brendan D. Price, Yingli Sun, and Xiaofeng Jiang
- Subjects
DNA repair ,DNA damage ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Lysine Acetyltransferase 5 ,Acetyltransferases ,medicine ,Humans ,CHEK1 ,Kinase activity ,DNA-PKcs ,Histone Acetyltransferases ,Binding Sites ,Multidisciplinary ,biology ,Tumor Suppressor Proteins ,Acetylation ,Histone acetyltransferase ,Biological Sciences ,medicine.disease ,Molecular biology ,DNA-Binding Proteins ,Enzyme Activation ,Protein Transport ,Mutation ,Ataxia-telangiectasia ,biology.protein ,DNA Damage ,HeLa Cells ,Protein Binding ,Signal Transduction - Abstract
The ataxia telangiectasia mutant (ATM) protein kinase regulates the cell's response to DNA damage through the phosphorylation of proteins involved in cell-cycle checkpoints and DNA repair. However, the signal-transduction pathway linking DNA strand breaks to activation of ATM's kinase activity is not clearly defined. Here, we demonstrate that DNA damage induces the rapid acetylation of ATM. This acetylation depends on the Tip60 histone acetyltransferase (HAT). Suppression of Tip60 blocks the activation of ATM's kinase activity and prevents the ATM-dependent phosphorylation of p53 and chk2. Further, inactivation of Tip60 sensitizes cells to ionizing radiation. ATM forms a stable complex with Tip60 through the conserved FATC domain of ATM. The interaction between ATM and Tip60 is not regulated in response to DNA damage. Instead, the HAT activity of the ATM–Tip60 complex is specifically activated by DNA damage. Furthermore, this activation of Tip60 by DNA damage and the recruitment of the ATM–Tip60 complex to sites of DNA damage is independent of ATM's kinase activity. The results demonstrate that the Tip60 HAT plays a key role in the activation of ATM's kinase activity in response to DNA damage.
- Published
- 2005
- Full Text
- View/download PDF
33. Stable siRNA-mediated silencing of ATM alters the transcriptional profile of HeLa cells
- Author
-
Gang Wang, G. Mike Makrigiorgos, Brendan D. Price, and Shujuan Chen
- Subjects
Transcription, Genetic ,Cell Survival ,DNA damage ,Cell ,Biophysics ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,Transfection ,Biochemistry ,Fluorescence ,Interferon ,Radiation, Ionizing ,medicine ,Humans ,Gene silencing ,Gene Silencing ,RNA, Small Interfering ,E2F ,Molecular Biology ,Integral membrane protein ,Oligonucleotide Array Sequence Analysis ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Tumor Suppressor Proteins ,Cell Biology ,Cell cycle ,medicine.disease ,Molecular biology ,Cell biology ,DNA-Binding Proteins ,medicine.anatomical_structure ,Gene Expression Regulation ,Ataxia-telangiectasia ,HeLa Cells ,medicine.drug - Abstract
The ATM protein, which is mutated in the inherited disease ataxia telangiectasia (AT), is a key regulator of the cells’ DNA damage response. AT cells also exhibit constitutive activation of transcriptional regulators such as p53, E2F, AP1, and NFκB. Inactivation of ATM may therefore alter the cells’ transcriptional profile. ATM expression in HeLa cells was silenced with siRNA expressed from a plasmid based vector, generating a stable cell line, HeLaATM601. HeLaATM601 cells displayed minimal levels of ATM protein and had a 10-fold increase in sensitivity to ionizing radiation. DNA microarray analysis demonstrated that 35 genes were upregulated and five genes were downregulated in HeLaATM601 cells. Genes upregulated in the absence of ATM included interferon-response proteins, cell cycle regulators, integral membrane proteins, and adhesion and extracellular matrix proteins. Using real-time PCR, these genes were also upregulated in cells derived from AT patients. Inactivation of the ATM protein therefore has a significant impact on the transcriptional profile of the cell.
- Published
- 2004
- Full Text
- View/download PDF
34. Ligation of a primer at a mutation: a method to detect low level mutations in DNA
- Author
-
Weihua Liu, Yuzhi Zhang, Sotirios Tetradis, Brendan D. Price, G. Mike Makrigiorgos, and Manjit Kaur
- Subjects
Lung Neoplasms ,DNA Ligases ,Health, Toxicology and Mutagenesis ,DNA Mutational Analysis ,Mutant ,Bone Neoplasms ,Adenocarcinoma ,Biology ,Toxicology ,Polymerase Chain Reaction ,law.invention ,chemistry.chemical_compound ,law ,Tumor Cells, Cultured ,Genetics ,Humans ,RNA, Messenger ,Ku Autoantigen ,Genetics (clinical) ,Polymerase chain reaction ,DNA Primers ,Osteosarcoma ,DNA Helicases ,Antigens, Nuclear ,Tenascin ,DNA, Neoplasm ,Genes, p53 ,Molecular biology ,DNA-Binding Proteins ,Restriction site ,genomic DNA ,Restriction enzyme ,chemistry ,Mutation ,Amplified fragment length polymorphism ,Restriction fragment length polymorphism ,Polymorphism, Restriction Fragment Length ,DNA - Abstract
Detection of low frequency mutations following exposure to mutagens or during the early stages of cancer development is instrumental for risk assessment and molecular diagnosis. We present a sensitive new method to detect trace levels of DNA mutations induced within a large excess of wild-type sequences. The method is based on mutation-induced generation of new restriction enzyme recognition sites. A DNA sequence is amplified from genomic DNA or cDNA using a high fidelity polymerase. The purified PCR product is digested with a restriction enzyme that recognizes the newly generated restriction site, partially dephosphorylated and ligated with an oligonucleotide at the position of the mutation. The ligated oligonucleotide is then utilized in two rounds of PCR to amplify the mutated DNA but not the wild-type allele that contains no restriction site. An A-->T polymorphism in mRNA (tenascin gene, A(2366)-->T, Asn-->Ile) and a G-->A polymorphism in genomic DNA (Ku gene, G(74582)-->A, Val-->Ile), both of which generate a restriction site for the enzyme SAU3A1, demonstrate the application. Eleven patient samples pre-characterized for the G(74582)-->A polymorphism in the repair gene Ku are used to demonstrate the reliability of this approach. This technique quantitatively detects the Ku G-->A polymorphism at a mutant frequency of 1.6x10(-6) relative to the wild-type allele. Mutations in p53 that are frequently induced by mutagens can readily be detected using the present method. As an example, using a second enzyme BbvI, a mutation frequently encountered in human cancers (G(14154)-->A mutation, p53 codon 245, Arg-->Gln) was detected in patient samples. The process does not require radioactivity, utilizes established procedures and overcomes several factors known to produce false positives in RFLP-based assays. The present amplification via primer ligation at the mutation (APRIL-ATM) has potential applications in the detection of mutagen-generated genetic alterations, early detection of tumor marker mutations in bodily discharges and the diagnosis of minimal residual disease.
- Published
- 2002
- Full Text
- View/download PDF
35. The tale of a tail: histone H4 acetylation and the repair of DNA breaks
- Author
-
Brendan D. Price, Surbhi Dhar, Ramya Parasuram, and Ozge Gursoy-Yuzugullu
- Subjects
0301 basic medicine ,DNA Repair ,biology ,DNA repair ,Acetylation ,Articles ,Molecular biology ,Chromatin ,General Biochemistry, Genetics and Molecular Biology ,Double Strand Break Repair ,Chromatin remodeling ,Histones ,03 medical and health sciences ,030104 developmental biology ,Histone ,Histone H2A ,biology.protein ,Animals ,Humans ,Histone code ,DNA Breaks, Double-Stranded ,General Agricultural and Biological Sciences ,Epigenomics - Abstract
The ability of cells to detect and repair DNA double-strand breaks (DSBs) within the complex architecture of the genome requires co-ordination between the DNA repair machinery and chromatin remodelling complexes. This co-ordination is essential to process damaged chromatin and create open chromatin structures which are required for repair. Initially, there is a PARP-dependent recruitment of repressors, including HP1 and several H3K9 methyltransferases, and exchange of histone H2A.Z by the NuA4-Tip60 complex. This creates repressive chromatin at the DSB in which the tail of histone H4 is bound to the acidic patch on the nucleosome surface. These repressor complexes are then removed, allowing rapid acetylation of the H4 tail by Tip60. H4 acetylation blocks interaction between the H4 tail and the acidic patch on adjacent nucleosomes, decreasing inter-nucleosomal interactions and creating open chromatin. Further, the H4 tail is now free to recruit proteins such as 53BP1 to DSBs, a process modulated by H4 acetylation, and provides binding sites for bromodomain proteins, including ZMYND8 and BRD4, which are important for DSB repair. Here, we will discuss how the H4 tail functions as a dynamic hub that can be programmed through acetylation to alter chromatin packing and recruit repair proteins to the break site. This article is part of the themed issue ‘Chromatin modifiers and remodellers in DNA repair and signalling’.
- Published
- 2017
- Full Text
- View/download PDF
36. Abstract B14: A novel role for BRD9 in regulating cellular growth and DNA damage response pathways
- Author
-
Farzin Gharadaghi, Alexis I. Cocozaki, Kelly Jacques, Sylvie Guichard, Caroline Vallaster, and Brendan D. Price
- Subjects
Cancer Research ,ARID1A ,DNA repair ,DNA damage ,Biology ,medicine.disease_cause ,Chromatin remodeling ,Chromatin ,Bromodomain ,Oncology ,SMARCA4 ,Cancer research ,medicine ,Carcinogenesis ,Molecular Biology - Abstract
Bromodomain containing protein BRD9 has been identified as a component of the SWI/SNF chromatin remodeling complex. Next generation sequencing studies have revealed that SWI/SNF is the most highly perturbed chromatin remodeling complex in cancer with subunit mutations present in approximately 20% of all tumor types. SWI/SNF has been implicated as both tumor suppressor and oncogenic driver in diverse contexts. For example, the catalytic subunit SMARCA4 functions as an oncogenic driver, essential for the maintenance of hematologic malignancies such as leukemia. In contrast, loss of function of the ATPase subunit SMARCA4 and the ARID1a subunit precipitate loss of tumor suppressor activity and are apparent in lung and ovarian subtypes. However, the role of BRD9 has not been clearly delineated. Clinical data indicates that testicular germ cell cancer (66%) and esophageal cancer (18%) bear a hemizygous deletion of BRD9 conferring HETLOSS status. Loss of BRD9 heterozygosity leading to enhanced tumorigenic potential is a hallmark of tumor suppressors. This highlights a novel role for this bromodomain protein in regulating the process of oncogenesis. Here we demonstrate shRNA-mediated knockdown of BRD9 enhances clonogenic potential of solid tumor cancer cell lines, while expression of wild-type BRD9 in a HETLOSS cell line abrogates colony formation. Collectively, our data suggests BRD9 functions as a regulator of cellular growth. We examined if SWI/SNF chromatin remodeling activity was dependent on BRD9 status and how this correlated with the growth phenotype. Nucleosome accessibility was significantly decreased when BRD9 was depleted indicating chromatin exists in a more compacted conformation. However, BRD9 knockdown does not result in alterations in assembly or stability of the SWI/SNF complex. Though loss of BRD9 does not perturb SWI/SNF dynamics, it may be required for complex recruitment to chromatin directly or by tethering through protein-protein interactions. Identification of novel BRD9-interacting proteins that target recruitment was carried out by proteomic survey. Analysis of the study revealed enrichment of proteins involved in DNA damage repair pathways. Using a double strand break reporter, we examined if BRD9 depletion perturbed homology-directed or non-homologous end joining repair pathways. Knockdown of BRD9 increased NHEJ-associated reporter activity but had subtle effect on homology-directed repair events. BRD9 colocalizes with γH2AX foci upon damage but this colocalization is lost upon depletion of BRD9. We propose that BRD9 is necessary for recruitment of SWI/SNF to sites of damage, to permit chromatin expansion and assembly of homology-directed repair factors. However, knockdown of BRD9 may shift the balance and favor interactions of non-homologous end joining pathway players to sites of damage, resulting in enhanced error-prone repair and ultimately leading to oncogenic transformation. Collectively, this work highlights a novel and previously unidentified role for BRD9 in DNA damage pathways and identifies a potential vulnerability in NHEJ dependence that may be therapeutically targeted. Citation Format: Caroline Vallaster, Farzin Gharadaghi, Alexis Cocozaki, Kelly Jacques, Brendan Price, Sylvie Guichard. A novel role for BRD9 in regulating cellular growth and DNA damage response pathways [abstract]. In: Proceedings of the AACR Special Conference on DNA Repair: Tumor Development and Therapeutic Response; 2016 Nov 2-5; Montreal, QC, Canada. Philadelphia (PA): AACR; Mol Cancer Res 2017;15(4_Suppl):Abstract nr B14.
- Published
- 2017
- Full Text
- View/download PDF
37. Dimer monomer transition and dimer re-formation play important role for ATM cellular function during DNA repair
- Author
-
Ye Xu, Minjie Zhang, Shuang Chang, Hao Meng, Yingli Sun, Caiyun Yang, Dong Wang, Brendan D. Price, Fengxia Du, and Xiaohua Li
- Subjects
DNA Repair ,Chemistry ,DNA damage ,DNA repair ,Dimer ,Autophosphorylation ,Biophysics ,Cell Biology ,Ataxia Telangiectasia Mutated Proteins ,Fibroblasts ,Biochemistry ,Article ,Phase Transition ,Cell Line ,Dephosphorylation ,chemistry.chemical_compound ,Phosphorylation ,Humans ,Protein kinase A ,Molecular Biology ,Dimerization ,DNA - Abstract
The ATM protein kinase, is a serine/threonine protein kinase that is recruited and activated by DNA double-strand breaks, mediates responses to ionizing radiation in mammalian cells. Here we show that ATM is held inactive in unirradiated cells as a dimer and phosphorylates the opposite strand of the dimer in response to DNA damage. Cellular irradiation induces rapid intermolecular autophosphorylation of serine 1981 that causes dimer dissociation and initiates cellular ATM kinase activity. ATM cannot phosphorylate the substrates when it could not undergo dimer monomer transition. After DNA repair, the active monomer will undergo dephosphorylation to form dimer again and dephosphorylation is critical for dimer re-formation. Our work reveals novel function of ATM dimer monomer transition and explains why ATM dimer monomer transition plays such important role for ATM cellular activity during DNA repair.
- Published
- 2014
38. DNA double-strand breaks promote methylation of histone H3 on lysine 9 and transient formation of repressive chromatin
- Author
-
Ye Xu, Chang Xu, Brendan D. Price, Ozge Gursoy-Yuzugullu, and Marina K. Ayrapetov
- Subjects
Ataxia Telangiectasia Mutated Proteins ,Biology ,Tripartite Motif-Containing Protein 28 ,Methylation ,Chromatin remodeling ,Lysine Acetyltransferase 5 ,Histone H4 ,Histones ,Histone H1 ,Histone H2A ,Histone methylation ,Histone code ,Humans ,DNA Breaks, Double-Stranded ,Phosphorylation ,Histone Acetyltransferases ,Multidisciplinary ,Lysine ,Methyltransferases ,Biological Sciences ,Chromatin Assembly and Disassembly ,Molecular biology ,Chromatin ,Cell biology ,Protein Structure, Tertiary ,Repressor Proteins ,HEK293 Cells ,Histone methyltransferase ,HeLa Cells - Abstract
Dynamic changes in histone modification are critical for regulating DNA double-strand break (DSB) repair. Activation of the Tip60 acetyltransferase by DSBs requires interaction of Tip60 with histone H3 methylated on lysine 9 (H3K9me3). However, how H3K9 methylation is regulated during DSB repair is not known. Here, we demonstrate that a complex containing kap-1, HP1, and the H3K9 methyltransferase suv39h1 is rapidly loaded onto the chromatin at DSBs. Suv39h1 methylates H3K9, facilitating loading of additional kap-1/HP1/suv39h1 through binding of HP1’s chromodomain to the nascent H3K9me3. This process initiates cycles of kap-1/HP1/suv39h1 loading and H3K9 methylation that facilitate spreading of H3K9me3 and kap-1/HP1/suv39h1 complexes for tens of kilobases away from the DSB. These domains of H3K9me3 function to activate the Tip60 acetyltransferase, allowing Tip60 to acetylate both ataxia telangiectasia-mutated (ATM) kinase and histone H4. Consequently, cells lacking suv39h1 display defective activation of Tip60 and ATM, decreased DSB repair, and increased radiosensitivity. Importantly, activated ATM rapidly phosphorylates kap-1, leading to release of the repressive kap-1/HP1/suv39h1 complex from the chromatin. ATM activation therefore functions as a negative feedback loop to remove repressive suv39h1 complexes at DSBs, which may limit DSB repair. Recruitment of kap-1/HP1/suv39h1 to DSBs therefore provides a mechanism for transiently increasing the levels of H3K9me3 in open chromatin domains that lack H3K9me3 and thereby promoting efficient activation of Tip60 and ATM in these regions. Further, transient formation of repressive chromatin may be critical for stabilizing the damaged chromatin and for remodeling the chromatin to create an efficient template for the DNA repair machinery.
- Published
- 2014
39. Activation of p53 transcriptional activity requires ATM's kinase domain and multiple N-terminal serine residues of p53
- Author
-
Gaetan A Turenne, Lareina Laflair, Brendan D. Price, and Proma Paul
- Subjects
Transcriptional Activation ,Antimetabolites, Antineoplastic ,Cancer Research ,Cell cycle checkpoint ,Transcription, Genetic ,DNA repair ,DNA damage ,Blotting, Western ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,Cell Line ,Bleomycin ,Transactivation ,Serine ,Genetics ,Humans ,Phosphorylation ,Kinase activity ,Molecular Biology ,Dose-Response Relationship, Drug ,Kinase ,Dose-Response Relationship, Radiation ,Genes, p53 ,Precipitin Tests ,Protein Structure, Tertiary ,Cell biology ,Enzyme Activation ,Cell killing ,Biochemistry ,Protein kinase domain ,Mutation ,Mutagenesis, Site-Directed ,Tumor Suppressor Protein p53 ,Gene Deletion ,DNA Damage - Abstract
The ATM protein kinase regulates the cell's response to DNA damage by regulating cell cycle checkpoints and DNA repair. ATM phosphorylates several proteins involved in the DNA-damage response, including p53. We have examined the mechanism by which ATM regulates p53's transcriptional activity. Here, we demonstrate that reintroduction of ATM into AT cells restores the activation of p53 by the radio-mimetic agent bleomycin. Further, p53 activation is lost when a kinase inactive ATM is used, or if the N-terminal of ATM is deleted. In addition, AT cells stably expressing ATM showed decreased sensitivity to Ionizing Radiation-induced cell killing, whereas cells expressing kinase inactive ATM or N-terminally deleted ATM were indistinguishable from AT cells. Finally, single point-mutations of serines 15, 20, 33 or 37 did not individually block the ATM-dependent activation of p53 transcriptional activity by bleomycin. However, double mutations of either serines 15 and 20 or serines 33 and 37 blocked the ability of ATM to activate p53. Our results indicate that the N-terminal of ATM and ATM's kinase activity are required for activation of p53's transcriptional activity and restoration of normal sensitivity to DNA damage. In addition, activation of p53 by ATM requires multiple serine residues in p53's transactivation domain.
- Published
- 2001
- Full Text
- View/download PDF
40. Constitutive activation of IκB kinase α and NF-κB in prostate cancer cells is inhibited by ibuprofen
- Author
-
Brendan D. Price, M Y Youmell, Stuart K. Calderwood, C. N. Coleman, and S T Palayoor
- Subjects
Cancer Research ,medicine.medical_specialty ,Kinase ,NF-κB ,IκB kinase ,Biology ,medicine.disease ,medicine.disease_cause ,Prostate cancer ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Cell culture ,Internal medicine ,LNCaP ,Genetics ,medicine ,Cancer research ,Protein kinase A ,Carcinogenesis ,Molecular Biology - Abstract
Apoptotic pathways controlled by the Rel/NF-κB family of transcription factors may regulate the response of cells to DNA damage. Here, we have examined the NF-κB status of several prostate tumor cell lines. In the androgen-independent prostate tumor cells PC-3 and DU-145, the DNA-binding activity of NF-κB was constitutively activated and IκB-α levels were decreased. In contrast, the androgen-sensitive prostate tumor cell line LNCaP had low levels of NF-κB which were upregulated following exposure to cytokines or DNA damage. The activity of the IκB-α kinase, IKKα, which mediates NF-κB activation, was also measured. In PC-3 cells, IKKα activity was constitutively active, whereas LNCaP cells had minimal IKKα activity that was activated by cytokines. The anti-inflammatory agent ibuprofen inhibited the constitutive activation of NF-κB and IKKα in PC-3 and DU-145 cells, and blocked stimulated activation of NF-κB in LNCaP cells. However, ibuprofen did not directly inhibit IκB-α kinase. The results demonstrate that NF-κB is constitutively activated in the hormone-insensitive prostate tumor cell lines PC-3 and DU-145, but not in the hormone responsive LNCaP cell line. The constitutive activation of NF-κB in prostate tumor cells may increase expression of anti-apoptotic proteins, thereby decreasing the effectiveness of anti-tumor therapy and contributing to the development of the malignant phenotype.
- Published
- 1999
- Full Text
- View/download PDF
41. Caffeine inhibits the checkpoint kinase ATM
- Author
-
Brendan D. Price, Alessandra Blasina, Gaetan A Turenne, and Clare H. McGowan
- Subjects
Time Factors ,DNA damage ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Biology ,Protein Serine-Threonine Kinases ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Wortmannin ,chemistry.chemical_compound ,Caffeine ,Humans ,Radiosensitivity ,Enzyme Inhibitors ,Phosphorylation ,Checkpoint Kinase 2 ,Adaptor Proteins, Signal Transducing ,Dose-Response Relationship, Drug ,Agricultural and Biological Sciences(all) ,Biochemistry, Genetics and Molecular Biology(all) ,Tumor Suppressor Proteins ,Cell Cycle ,Cell cycle ,G2-M DNA damage checkpoint ,Phosphoproteins ,Precipitin Tests ,Androstadienes ,DNA-Binding Proteins ,Biochemistry ,chemistry ,Cell culture ,Cancer research ,General Agricultural and Biological Sciences ,Carrier Proteins ,Protein Kinases ,DNA Damage ,HeLa Cells - Abstract
The basis of many anti-cancer therapies is the use of genotoxic agents that damage DNA and thus kill dividing cells. Agents that cause cells to override the DNA-damage checkpoint are predicted to sensitize cells to killing by genotoxic agents. They have therefore been sought as adjuncts in radiation therapy and chemotherapy. One such compound, caffeine, uncouples cell-cycle progression from the replication and repair of DNA [1] [2]. Caffeine therefore servers as a model compound in establishing the principle that agents that override DNA-damage checkpoints can be used to sensitize cells to the killing effects of genotoxic drugs [3]. But despite more than 20 years of use, the molecular mechanisms by which caffeine affects the cell cycle and checkpoint responses have not been identified. We investigated the effects of caffeine on the G2/M DNA-damage checkpoint in human cells. We report that the radiation-induced activation of the kinase Cds1 [4] (also known as Chk2 [5]) is inhibited by caffeine in vivo and that ATM kinase activity is directly inhibited by caffeine in vitro. Inhibition of ATM provides a molecular explanation of the attenuation of DNA-damage checkpoint responses and for the increased radiosensitivity of caffeine-treated cells [6] [7] [8].
- Published
- 1999
- Full Text
- View/download PDF
42. An essential role of NFκB in tyrosine kinase signaling of p38 MAP kinase regulation of myocardial adaptation to ischemia
- Author
-
Brendan D. Price, Nilanjana Maulik, Dipak K. Das, and Motoaki Sato
- Subjects
Pyridines ,Myocardial Ischemia ,Signal transduction ,p38 Mitogen-Activated Protein Kinases ,Biochemistry ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Ischemia ,Structural Biology ,Malondialdehyde ,ASK1 ,Creatine Kinase ,Imidazoles ,NF-kappa B ,Heart ,Protein-Tyrosine Kinases ,Adaptation, Physiological ,Genistein ,Cell biology ,Mitogen associated protein kinase ,Mitogen-activated protein kinase ,Mitogen-Activated Protein Kinases ,Tyrosine kinase ,medicine.medical_specialty ,SB 203580 ,p38 mitogen-activated protein kinases ,Biophysics ,Myocardial Reperfusion Injury ,In Vitro Techniques ,Biology ,Internal medicine ,Genetics ,medicine ,Animals ,Nuclear factor κB ,Adaptation ,Molecular Biology ,Cell Biology ,medicine.disease ,Rats ,Endocrinology ,chemistry ,Oxidative stress ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Ischemic preconditioning ,Peptides - Abstract
We have recently demonstrated that myocardial adaptation to ischemia triggers a tyrosine kinase regulated signaling pathway leading to the translocation and activation of p38 MAP kinase and MAPKAP kinase 2. Since oxidative stress is developed during ischemic adaptation and since free radicals have recently been shown to function as an intracellular signaling agent leading to the activation of nuclear transcription factor, NFkappaB, we examined whether NFkappaB was involved in the ischemic adaptation process. Isolated perfused rat hearts were adapted to ischemic stress by repeated ischemia and reperfusion. Hearts were pretreated with genistein to block tyrosine kinase while SB 203580 was used to inhibit p38 MAP kinases. Ischemic adaptation was associated with the nuclear translocation and activation of NFkappaB which was significantly blocked by both genistein and SB 203580. The ischemically adapted hearts were more resistant to ischemic reperfusion injury as evidenced by better function recovery and less tissue injury during post-ischemic reperfusion. Ischemic adaptation developed oxidative stress which was reflected by increased malonaldehyde formation. A synthetic peptide containing a cell membrane-permeable motif and nuclear sequence, SN 50, which blocked nuclear translocation of NFkappaB during ischemic adaptation, significantly inhibited the beneficial effects of adaptation on functional recovery and tissue injury. In concert, SN 50 reduced the oxidative stress developed in the adapted myocardium. These results demonstrate that p38 MAP kinase might be upstream of NFkappaB which plays a role in ischemic preconditioning of heart.
- Published
- 1998
- Full Text
- View/download PDF
43. The DNA-Dependent Protein Kinase Participates in the Activation of NFκB Following DNA Damage
- Author
-
Subimal Basu, Brendan D. Price, Matthew B. Youmell, and Kenneth R. Rosenzweig
- Subjects
DNA damage ,medicine.medical_treatment ,Biophysics ,DNA-Activated Protein Kinase ,Protein Serine-Threonine Kinases ,Biology ,Biochemistry ,Wortmannin ,chemistry.chemical_compound ,NF-KappaB Inhibitor alpha ,Tumor Cells, Cultured ,medicine ,Humans ,Phosphorylation ,Nuclear protein ,Molecular Biology ,Transcription factor ,Etoposide ,Tumor Necrosis Factor-alpha ,Kinase ,NF-kappa B ,Nuclear Proteins ,Glioma ,Cell Biology ,Molecular biology ,Androstadienes ,DNA-Binding Proteins ,Cytokine ,chemistry ,Gamma Rays ,I-kappa B Proteins ,Tumor necrosis factor alpha ,DNA Damage ,HeLa Cells - Abstract
The NFkB transcription factor is activated by diverse stimuli, including Ionizing Radiation (IR) and the cytokine TNF alpha. The role of DNA-PK, a protein kinase involved in the response to DNA damage, in the activation of NF kappa B by IR and TNF alpha was examined. In M059K cells, which express DNA-PK, NF kappa B was activated by both TNF alpha and IR. In M059J cells, which do not express DNA-PK, IR did not activate NF kappa B, whereas TNF alpha induction of NF kappa B was still observed. In HeLa cells, wortmannin, an inhibitor of DNA-PK, blocked the induction of NF kappa B by IR but not by TNF alpha. DNA-PK also phosphorylated the NF kappa B inhibitory proteins IkB-alpha and IkB-beta in vitro, and deletion analysis demonstrated that DNA-PK phosphorylates 2 distinct regions of IkB-beta. These results indicate that DNA-PK participates in the activation of NF kappa B by IR but not by TNF alpha.
- Published
- 1998
- Full Text
- View/download PDF
44. Regulation of the p53 Protein by Protein Kinase Cα and Protein Kinase Cζ
- Author
-
Brendan D. Price, Subimal Basu, Matthew B. Youmell, and Sonia J. Park
- Subjects
MAP kinase kinase kinase ,Chemistry ,Mutagenesis ,Biophysics ,Cell Biology ,Mitogen-activated protein kinase kinase ,Biochemistry ,Molecular biology ,MAP2K7 ,Serine ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Protein kinase C - Abstract
The C-terminal of p53 (amino-acids 368-383) represses the DNA binding activity of p53. In vitro, phosphorylation of this region by Protein Kinase C (PKC) is associated with increased DNA binding activity. However, whether PKC can directly modulate p53 function in vivo is not known. Here, we demonstrate that cotransfection of p53 with either PKCα or PKCζ increases p53's transcriptional activity. Mutagenesis of p53 indicates that serine 371 is the major site for phosphorylation by PKCα in vitro. Mutation of serine 371 caused a small decline in p53 activation by PKCα and PKCζ. However, the alternatively spliced murine p53, which lacks the PKC phosphorylation sites, still demonstrated increased transcriptional activation when cotransfected with either PKCα or PKCζ. The results indicate that phosphorylation of p53 by PKC in vitro does not correlate with the ability of PKC to upregulate p53's transcriptional activity in vivo.
- Published
- 1998
- Full Text
- View/download PDF
45. Sequential Phosphorylation by Mitogen-activated Protein Kinase and Glycogen Synthase Kinase 3 Represses Transcriptional Activation by Heat Shock Factor-1
- Author
-
Brendan D. Price, Boyang Chu, Stuart K. Calderwood, Fabrice Soncin, and Mary Ann Stevenson
- Subjects
MAPK/ERK pathway ,endocrine system ,Transcription, Genetic ,MAPK cascade ,Peptide Mapping ,Biochemistry ,HSPA4 ,Glycogen Synthase Kinase 3 ,Phosphoserine ,Structure-Activity Relationship ,Heat Shock Transcription Factors ,GSK-3 ,Heat shock protein ,Humans ,HSP70 Heat-Shock Proteins ,Phosphorylation ,Heat shock ,Promoter Regions, Genetic ,HSF1 ,Molecular Biology ,Mitogen-Activated Protein Kinase 3 ,biology ,Chemistry ,Glycogen Synthase Kinases ,Cell Biology ,Molecular biology ,DNA-Binding Proteins ,Gene Expression Regulation ,Mitogen-activated protein kinase ,Calcium-Calmodulin-Dependent Protein Kinases ,biology.protein ,Mitogen-Activated Protein Kinases ,Signal Transduction ,Transcription Factors - Abstract
Mammalian heat shock genes are regulated at the transcriptional level by heat shock factor-1 (HSF-1), a sequence-specific transcription factor. We have examined the role of serine phosphorylation of HSF-1 in the regulation of heat shock gene transcription. Our experiments show that mitogen-activated protein kinases (MAPKs) of the ERK-1 family phosphorylate HSF-1 on serine residues and repress the transcriptional activation of the heat shock protein 70B (HSP70B) promoter by HSF-1 in vivo. These effects of MAPK are transmitted through a specific serine residue (Ser-303) located in a proline-rich sequence within the transcriptional regulatory domain of human HSF-1. However, despite the importance of Ser-303 in transmitting the signal from the MAPK cascade to HSP70 transcription, there was no evidence that Ser-303 could be phosphorylated by MAPK in vitro, although an adjacent residue (Ser-307) was avidly phosphorylated by MAPK. Further studies revealed that Ser-303 is phosphorylated by glycogen synthase kinase 3 (GSK3) through a mechanism dependent on primary phosphorylation of Ser-307 by MAPK. Secondary phosphorylation of Ser-303 by GSK3 may thus repress the activity of HSF-1, and its requirement for priming by MAPK phosphorylation of Ser-307 provides a potential link between the MAPK cascade and HSF-1. Our experiments thus indicate that MAPK is a potent inhibitor of HSF-1 function and may be involved in repressing the heat shock response during normal growth and development and deactivating the heat shock response during recovery from stress.
- Published
- 1996
- Full Text
- View/download PDF
46. Mechanistic links between ATM and histone methylation codes during DNA repair
- Author
-
Ye, Xu, Chang, Xu, and Brendan D, Price
- Subjects
DNA-Binding Proteins ,Histone Code ,Histones ,DNA Repair ,Neoplasms ,Tumor Suppressor Proteins ,Animals ,Humans ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Methylation - Abstract
The ataxia telangiectasia-mutated (ATM) protein kinase is the master regulator of the DNA double-strand break (DSB) repair pathway. The activation of ATM involves its recruitment to the DSB through interaction with the mre11-rad50-nbs1 complex, followed by the acetylation of ATM by the Tip60 acetyltransferase. This acetylation of ATM within its regulatory domain is essential for activating ATM's kinase activity. Further work has now revealed that Tip60 is activated through direct interaction between Tip60's chromodomain and histone H3 trimethylated on lysine 9 (H3K9me3). The loading of Tip60 onto the chromatin at DSBs therefore represents the primary mechanism for activation of Tip60's acetyltransferase activity in response to DNA damage. The ability of H3K9me3 at DSBs to regulate the activity of Tip60 and the subsequent activation of ATM emphasizes the crucial role played by chromatin architecture in regulating DSB repair. Further, histone methylation and chromatin structure are disrupted in human cancers, implying that altered chromatin structure in tumor cells may impact DSB repair, increasing genomic instability and contributing to the progression of cancer.
- Published
- 2012
47. Mechanistic Links Between ATM and Histone Methylation Codes During DNA Repair
- Author
-
Ye Xu, Brendan D. Price, and Chang Xu
- Subjects
enzymes and coenzymes (carbohydrates) ,Histone H3 ,DNA repair ,Histone methyltransferase ,Histone H2A ,Histone methylation ,Cancer research ,Histone code ,biological phenomena, cell phenomena, and immunity ,Biology ,Chromatin remodeling ,Chromatin - Abstract
The ataxia telangiectasia-mutated (ATM) protein kinase is the master regulator of the DNA double-strand break (DSB) repair pathway. The activation of ATM involves its recruitment to the DSB through interaction with the mre11-rad50-nbs1 complex, followed by the acetylation of ATM by the Tip60 acetyltransferase. This acetylation of ATM within its regulatory domain is essential for activating ATM's kinase activity. Further work has now revealed that Tip60 is activated through direct interaction between Tip60's chromodomain and histone H3 trimethylated on lysine 9 (H3K9me3). The loading of Tip60 onto the chromatin at DSBs therefore represents the primary mechanism for activation of Tip60's acetyltransferase activity in response to DNA damage. The ability of H3K9me3 at DSBs to regulate the activity of Tip60 and the subsequent activation of ATM emphasizes the crucial role played by chromatin architecture in regulating DSB repair. Further, histone methylation and chromatin structure are disrupted in human cancers, implying that altered chromatin structure in tumor cells may impact DSB repair, increasing genomic instability and contributing to the progression of cancer.
- Published
- 2012
- Full Text
- View/download PDF
48. Activation of Hif1α by the prolylhydroxylase inhibitor dimethyoxalyglycine decreases radiosensitivity
- Author
-
Kalindi Parmar, Chang Xu, Brendan D. Price, Yingli Sun, Alan D. D'Andrea, Marina K. Ayrapetov, and Kaya Zhu
- Subjects
Radioprotection ,Cell ,lcsh:Medicine ,Autoantigens ,Radiation Tolerance ,Biochemistry ,Mice ,Molecular cell biology ,0302 clinical medicine ,Transcriptional regulation ,Enzyme Inhibitors ,lcsh:Science ,Cellular Stress Responses ,0303 health sciences ,Multidisciplinary ,Amino Acids, Dicarboxylic ,Neoplasm Proteins ,Up-Regulation ,Cell biology ,Nucleic acids ,medicine.anatomical_structure ,Oncology ,Hypoxia-inducible factors ,030220 oncology & carcinogenesis ,DNA methylation ,Medicine ,Mi-2 Nucleosome Remodeling and Deacetylase Complex ,Research Article ,DNA damage ,Radiation Biophysics ,DNA transcription ,Procollagen-Proline Dioxygenase ,Biophysics ,DNA repair ,Radiation-Protective Agents ,Biology ,03 medical and health sciences ,Histone H3 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,RNA, Messenger ,Radiosensitivity ,Transcription factor ,030304 developmental biology ,Radiotherapy ,lcsh:R ,Radiobiology ,DNA ,Fibroblasts ,Hypoxia-Inducible Factor 1, alpha Subunit ,Molecular biology ,HEK293 Cells ,lcsh:Q ,Gene expression - Abstract
Hypoxia inducible factor 1α (Hif1α) is a stress responsive transcription factor, which regulates the expression of genes required for adaption to hypoxia. Hif1α is normally hydroxylated by an oxygen-dependent prolylhydroxylase, leading to degradation and clearance of Hif1α from the cell. Under hypoxic conditions, the activity of the prolylhydroxylase is reduced and Hif1α accumulates. Hif1α is also constitutively expressed in tumor cells, where it is associated with resistance to ionizing radiation. Activation of the Hif1α transcriptional regulatory pathway may therefore function to protect normal cells from DNA damage caused by ionizing radiation. Here, we utilized the prolylhydroxylase inhibitor dimethyloxalylglycine (DMOG) to elevate Hif1α levels in mouse embryonic fibroblasts (MEFs) to determine if DMOG could function as a radioprotector. The results demonstrate that DMOG increased Hif1α protein levels and decreased the sensitivity of MEFs to ionizing radiation. Further, the ability of DMOG to function as a radioprotector required Hif1α, indicating a key role for Hif1α's transcriptional activity. DMOG also induced the Hif1α -dependent accumulation of several DNA damage response proteins, including CHD4 and MTA3 (sub-units of the NuRD deacetylase complex) and the Suv39h1 histone H3 methyltransferase. Depletion of Suv39h1, but not CHD4 or MTA3, reduced the ability of DMOG to protect cells from radiation damage, implicating increased histone H3 methylation in the radioprotection of cells. Finally, treatment of mice with DMOG prior to total body irradiation resulted in significant radioprotection of the mice, demonstrating the utility of DMOG and related prolylhydroxylase inhibitors to protect whole organisms from ionizing radiation. Activation of Hif1α through prolylhydroxylase inhibition therefore identifies a new pathway for the development of novel radiation protectors.
- Published
- 2011
49. Activation of phospholipase C by heat shock requires GTP analogs and is resistant to pertussis toxin
- Author
-
Mary Ann Stevenson, Brendan D. Price, and Stuart K. Calderwood
- Subjects
Hot Temperature ,GTP' ,Physiology ,G protein ,Clinical Biochemistry ,CHO Cells ,Inositol 1,4,5-Trisphosphate ,Phospholipase ,Biology ,Pertussis toxin ,Mice ,GTP-Binding Proteins ,Cricetinae ,Heat shock protein ,Animals ,Humans ,Virulence Factors, Bordetella ,Heat shock ,Phospholipase C ,Thrombin ,3T3 Cells ,Cell Biology ,Cell biology ,Enzyme Activation ,Pertussis Toxin ,Biochemistry ,Type C Phospholipases ,Guanosine Triphosphate ,Signal transduction ,HeLa Cells ,Signal Transduction - Abstract
The heat shock response in mammals consists of a complex array of intracellular reactions initiated by stress, although its regulation is poorly understood. We have investigated the role of transmembrane signal transduction in the response, examining mechanisms involved in the activation of phospholipase C (PLC) by heat shock. In rodent fibroblasts permeabilized with digitonin, heat shock and receptor-mediated PLC activity exhibited a strict GTP analog dependency. This indicates that heat shock-mediated phopholipase activation, in common with receptor mediated stimulation, does not involve direct effects on the phospholipases and suggests the participation of GTP binding (G) proteins in the activation process. When cells were treated with the inhibitor pertussis toxin (PTX), the phospholipases retained their inducibility by heat shock, but became refractory to thrombin treatment, indicating that heat shock may influence PLC activity through a distinct population of G proteins compared to thrombin. The data seem to exclude a role for PTX sensitive G proteins in the production of IP3 after heating and suggest a pathway involving the direct thermal activation of the Gq class of G proteins, which are coupled to the PLCβ1 isoform. © 1993 Wiley-Liss, Inc.
- Published
- 1993
- Full Text
- View/download PDF
50. Radiosensitization of mammary carcinoma cells by telomere homolog oligonucleotide pretreatment
- Author
-
Monique C Cunin, Baizheng Song, Stuart K. Calderwood, Barbara A. Gilchrest, Mark S. Eller, Brendan D. Price, Jianlin Gong, and Desheng Weng
- Subjects
Radiation-Sensitizing Agents ,Oligonucleotides ,Apoptosis ,Mammary Neoplasms, Animal ,Biology ,medicine.disease_cause ,Radiation Tolerance ,Histones ,Mice ,In vivo ,Cell Line, Tumor ,Radiation, Ionizing ,In Situ Nick-End Labeling ,medicine ,Animals ,Radiosensitivity ,Phosphorylation ,Clonogenic assay ,Cellular Senescence ,Cell Proliferation ,Medicine(all) ,Mammary tumor ,TUNEL assay ,Galactosides ,Telomere ,Combined Modality Therapy ,Molecular biology ,Mice, Inbred C57BL ,Terminal deoxynucleotidyl transferase ,Female ,Comet Assay ,Carcinogenesis ,Cell aging ,DNA Damage ,Research Article - Abstract
Introduction Ionizing radiation (IR) is a widely used approach to cancer therapy, ranking second only to surgery in rate of utilization. Responses of cancer patients to radiotherapy depend in part on the intrinsic radiosensitivity of the tumor cells. Thus, promoting tumor cell sensitivity to IR could significantly enhance the treatment outcome and quality of life for patients. Methods Mammary tumor cells were treated by a 16-base phosphodiester-linked oligonucleotide homologous to the telomere G-rich sequence TTAGGG (T-oligo: GGTTAGGTGTAGGTTT) or a control-oligo (the partial complement, TAACCCTAACCCTAAC) followed by IR. The inhibition of tumor cell growth in vitro was assessed by cell counting and clonogenic cell survival assay. The tumorigenesis of tumor cells after various treatments was measured by tumor growth in mice. The mechanism underlying the radiosensitization by T-oligo was explored by immunofluorescent determination of phosphorylated histone H2AX (γH2AX) foci, β-galactosidase staining, comet and Terminal deoxynucleotidyl transferase dUTP Nick End Labeling (TUNEL) assays. The efficacy of the combined treatment was assessed in a spontaneous murine mammary tumor model. Results Pretreatment of tumor cells with T-oligo for 24 hours in vitro enhanced both senescence and apoptosis of irradiated tumor cells and reduced clonogenic potential. Radiosensitization by T-oligo was associated with increased formation and/or delayed resolution of γH2AX DNA damage foci and fragmented DNA. T-oligo also caused radiosensitization in two in vivo mammary tumor models. Indeed, combined T-oligo and IR-treatment in vivo led to a substantial reduction in tumor growth. Of further significance, treatment with T-oligo and IR led to synergistic inhibition of the growth of spontaneous mammary carcinomas. Despite these profound antitumor properties, T-oligo and IR caused no detectable side effects under our experimental conditions. Conclusions Pretreatment with T-oligo sensitizes mammary tumor cells to radiation in both in vitro and in vivo settings with minimal or no normal tissue side effects.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.